CA2488716A1 - Intracellular delivery of biological effectors - Google Patents

Intracellular delivery of biological effectors Download PDF

Info

Publication number
CA2488716A1
CA2488716A1 CA002488716A CA2488716A CA2488716A1 CA 2488716 A1 CA2488716 A1 CA 2488716A1 CA 002488716 A CA002488716 A CA 002488716A CA 2488716 A CA2488716 A CA 2488716A CA 2488716 A1 CA2488716 A1 CA 2488716A1
Authority
CA
Canada
Prior art keywords
peptide
transporter
effector
cell
unit
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002488716A
Other languages
French (fr)
Inventor
Christophe Bonny
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Xigen SA
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2488716A1 publication Critical patent/CA2488716A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1016Tetrapeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/04General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length on carriers
    • C07K1/047Simultaneous synthesis of different peptide species; Peptide libraries
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1013Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing O or S as heteroatoms, e.g. Cys, Ser
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1019Tetrapeptides with the first amino acid being basic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1021Tetrapeptides with the first amino acid being acidic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S530/00Chemistry: natural resins or derivatives; peptides or proteins; lignins or reaction products thereof
    • Y10S530/81Carrier - bound or immobilized peptides or proteins and the preparation thereof, e.g. biological cell or cell fragment as carrier

Abstract

The invention relates to sequences of amino acids with the capacity to facilitate transport of an effector across a biological membrane. More specifically, the present invention relates to novel peptide transporters th at specifically target certain cell types for the intracellular delivery of dru gs and therapeutic agents.

Description

INTRACELLULAR DELIVERY OF BIOLOGICAL EFFECTORS
TECFINICAL FIELD OF THE INVENTION
The invention relates to the field of molecular biology.
BACKGROUND OF THE INVENTION
Techniques enabling the efficient transfer of a substance of interest from the external medium into cells, and particularly into cellular nuclei, are of considerable interest in the field of biotechnology. Such techniques may be useful for protein or peptide production, for regulation , of gene expression, for analysis of intracellular signaling channels and for analysis of the effect of transporting a variety of different substances into a cell (or cell nucleus). Currently available techniques are often limited by the inability of a transfer vector to transfer biologically active substances into the cytoplasm (or nuclei) of cells in the host to be treated without affecting the host genome or altering the biological properties of the active substance.
Previously, transduction of large active substances into cells was limited by the size and biochemical properties of the substance being transported. However, recent progress in this field has been achieved with the use of transporter peptides or proteins. For example, Schwarze and colleagues described a 10-mer peptide derived from HIV, TAT48_5~, which is capable of transporting conjugated peptides or proteins intracellularly into tissues and across the blood-~ brain barrier. Schwarze et al., Science 285:1573 (1999). These conjugated peptides or proteins are thought to be internalized by cells via a protein transduction process that does not involve endocytosis. This discovery opened a new methodology for biomedical research and for direct delivery of drugs into patients. However, one important limitation of this approach is the lack of cellular specificity for these types of transporters. Thus, adverse side effects from interaction with normal, non-target tissues limit the usefulness of many of these non-specific transporter peptides. Such a limitation is particularly problematic in the treatment of chronic diseases such as diabetes.

A need remains in the art for an efficient, safe compositions and methods for specifically targeting various cell types for the intracellular delivery of drugs and therapeutic agents via peptide transport.
SUMMARY OF THE INVENTION
The present invention provides transporter peptides which are capable of translocating across a biological membrane. The invention also relates to methods of using such transporter peptides to translocate an effector across a biological membrane. The transporter peptides can be monomeric or multimeric.
In one aspect, the invention involves transporter peptides having at least one amino acid sequence selected from: (XmRXoRX"); (XmRRRX"); (XmRWXRX"); and (XmR~~tRXn), where X
is a non-basic amino acid; m is an integer from zero to fourteen; n is an integer, independent of m, between zero and fourteen; o is an integer, independent of m and n, between zero and five;
and wherein the transporter peptide is capable of translocating across a biological membrane.
In one embodiment, the invention provides a transporter peptide having the amino acid sequence R-X-X-R. In other embodiments, the invention provides a transporter peptide having an amino acid sequence of any one of SEQ ID NOS: 1-34. In various other embodiments, the transporter peptides are derived from protein convertase ligands. In still other embodiments, the transporter peptides are derived from protein convertase cleavage sites.
In another aspect, the invention involves multimeric transporter peptides including two, three, four, five, six, seven, eight or more monomeric transporter peptides wherein each monomeric transporter peptide has at least one amino acid sequence selected from:
(Xm~o~n)e (Xmn); (Xm~'~n); and (XmRXRRXn), where X is a non-basic amino acid; m is an integer from zero to fourteen; n is an integer, independent of m, between zero and fourteen; o is an integer, independent of m and n, between zero and five; and wherein the transporter peptide is capable of translocating across a biological membrane.
The individual monomeric transporter peptides within a multimeric transporter peptide can be the same or different.
In another embodiment, the invention provides a multimeric transporter peptide having the amino acid sequence R-X-X-R. In other embodiments, the invention provides a multimeric transporter peptide having an amino acid sequence of any one of SEQ ID NOS: 1-34. In various other embodiments, the multimeric transporter peptides are derived from protein convertase ligands. In still other embodiments, the multimeric transporter peptides are derived from protein convertase cleavage sites.
In some embodiments, the multimeric transporter peptides include a spacer moiety and a linker moiety. Preferably, the spacer moiety and linker moiety are flexible, amphiphilic, non-immunogenic and unsusceptible to proteases. The linker can be, for example, polyethylene glycol.
In other embodiments, the monomeric or polymeric transporter peptide can include a reporter group. As used herein, a "reporter group" is any group capable of detection. Non-limiting examples include radioactive isotopes, fluorescent moieties, phosphorescent moieties, chemiluminescent moieties, and quantum dots. Other reporter groups include biotin, cysteine, histidine, haemagglutinin, myc or flag tags.
In some embodiments, transport peptides of the invention are represented by Formula I:
[P]~ [Spacer]X Core-[Linker]y-[Reporter]Z
where P is a peptide, falling within the consensus motifs (X",RXoRXn)i (Xmn)i (XmRRXRX"); and (XmI2~~RRXn), as described above. For example, the peptide can include the sequence Xl-X2x3-X4, where Xl and X4 can be R or K and Xl and X4 can be any amino acid. In one embodiment, the invention provides a multimeric transporter with at least one peptide having the amino acid sequence R-X-X-R. In other embodiments, the invention provides a multimeric transporter peptide with at least one peptide having an amino acid sequence of any one of SEQ
117 NOS: 1-34. In various embodiments, the transporter peptides are derived from protein convertase ligands. In still other embodiments, the transporter peptides are derived from protein convertase cleavage sites. The number of peptides in a given transporter, represented by v, is an integer. In monomeric transporter, v is one. In a multimeric transporter, v is an integer from two to eight or more. In any given multimeric conjugate, each of the peptides may be the same as or different from each other.
In a monomeric transporter construct, the core may be present or absent.
A spacer may be present or absent between the peptide and core, thus x is either zero or one. Preferred spacers are flexible, amphiphilic, non-immunogenic and unsusceptible to proteases, and include polyethylene glycols (PEG) such as succinimidyl-PEG
(succ-peg).
As used herein, a "core" is a structure that serves as an attachment point for linker, spacer, and reporter groups. The core also serves to connect the peptides within a multimeric transporter. In some embodiments, the core is branched so that many peptides can be attached in the same transporter unit. Typically, a core will have many functionalities available for attachment so that the various different moieties (the peptides and spacers, linkers, and reporter, if present) of the multimeric construct can be fused to the core, thereby generating one molecule.
The contemplated core moieties include the polylysine (K) cores described in detail below and also include those structures having a built-in linker. The chosen core determines the number of peptides present within a given conjugate. Typical cores include: C4-KKK-K(succ-peg-S)-amide;
C-GGG -[K(C)]3-K(succ-peg-S)-amide; (NHZOCH2C0)4-KZK-K(succ-peg)-amide;
(NHaOCH2C0)8-K4KzK-K(GGG)-amide.
In the multimeric transporter construct, the linker and reporter groups can be present or absent, thus y and z are, independently, one or zero in Formula I.
As used herein, a "transporter peptide" is a peptide that facilitates the translocation of a substance across a biological membrane. Unless otherwise specified, or inaccurate in the context used, reference to a transporter peptide encompasses a rnonomeric or multimeric transporter peptide.
In some embodiments, the transporter peptide is fused to one or more effectors. The "effector" can be any suitable molecule, including DNA, RNA, a protein, a peptide, or a pharmaceutically active agent, such as, for example, a toxin, an antibiotic, an antipathogenic agent, an antigen, an antibody, an antibody fragment, an immunomodulator, an enzyme, or a therapeutic agent. When more than one effector is present, the effectors can be the same or different.
The term "fusion" or "fused" is meant to include all such specific interactions that result in two or more molecules showing a preference for one another relative to some third molecule.
This includes processes such as covalent, ionic, hydrophobic, and hydrogen bonding, but does not include non-specific associations such as solvent preferences.
The multimeric conjugates of the invention can be represented by Formula (II):
([P]~ [Spacer]X Core-[Linker]y-[Reporter]Z)~E
where P, Core, Spacer, Linker, Reporter, v~ X, y, and z are as described above and E is an effector. In a covalently fused conjugate, the effector can be attached to the core, to the linker, or to both, as the laws of chemistry apply.
As used herein, "conjugate" or "conjugation" refer to any type of interaction enabling a physical association between an effector and a monomeric or multimeric transporter peptide.
The association may be covalent or non-covalent in nature, and it must be sufficiently strong so that the conjugate does not disassociate before or during cellular translocation. Conjugation may be achieved using any chemical, biochemical, enzymatic or genetic coupling known to those skilled in the art. The effector of interest may be coupled or fused to the N-terminal or C-terminal end of the transporter peptide. In some embodiments, the effector is coupled or fused to a linker group, directly to the core, or to a reporter group.
In various embodiments, the monomeric and/or multimeric transporter peptides can be less than fifty (50), less than twenty-five (25), or less than fifteen (15) amino acids in length.
In further embodiments, translocation occurs within pancreatic B-cells, hepatocytes, colon cells, muscle cells and/or lung cells.
In another embodiment, the invention involves a method of translocating a transporter peptide across a biological membrane. For example, monomeric or multimeric peptides containing one or more of the sequences of SEQ m NOS: 1-6 can be translocated across a membrane of pancreatic B-cells; monomeric or multimeric peptides containing one or more of the sequences of SEQ m NOS: 7-10 can be translocated across a membrane of hepatocytes;
monomeric or multimeric peptides containing one or more peptides of SEQ m NO:11 can be translocated across a membrane of colon cells; monomeric or multimeric peptides containing one or more of the sequences of SEQ m NOS: 12 -20 can be translocated across a membrane of muscle cells; and monomeric or multimeric peptides containing one or more of the sequences of SEQ m NOS: 21-34 can be translocated across a membrane of lung cells.
In yet another embodiment, the invention involves a transporter unit that is a monomeric or multimeric transporter peptide conjugated to one or more effectors. In various other embodiments, the effector may be a nucleic acid, a peptide, or a pharmaceutically active agent.
In still a further embodiment, the invention includes a method of producing a monomeric or multimeric translocatable conjugate between a transporter.peptide and one or more effectors, thereby forming a transporter peptide-effector conjugate.
In another embodiment, the invention includes a method of translocating one or more effectors into the cytoplasm and/or nucleus of a eukaryotic cell, whereby the effector is conjugated to a monomeric or multimeric transporter peptide and introduced into the eukaryotic cell. For example, the transporter peptide-effector conjugate can be introduced into the cell by incubating a cell culture in the presence of the conjugate or by injecting the conjugate into the cell.
In various other embodiments, the invention includes a method of increasing the cellular concentration of an effector within a eukaryotic cell, whereby an effector is conjugated to a monomeric or multimeric transporter peptide and incubated in the presence of a cell under conditions promoting active metabolism of the cell. A preferred embodiment of the invention includes use of a human cell as the eukaryotic cell.
In further embodiments, the invention includes a pharmaceutical composition containing a therapeutically or prophylactically effective amount of a monomeric or multimeric transporter unit and a pharmaceutically acceptable Garner.
1 S Preferred "pharmaceutical compositions" include tablets and gelatin capsules comprising the active ingredient together with a) diluents, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose andlor glycine; b) lubricants, e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone; if desired d) disintegrants, e.g., starches, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or e) absorbents, colorants, flavors and sweeteners. Injectable compositions are preferably aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions. The compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers.
In addition, they may also contain other therapeutically valuable substances. The compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1 to 75%, preferably about 1 to 50%, of the active ingredient.
In yet still further embodiments, the invention includes a kit in which one or more containers containing a therapeutically or prophylactically effective amount of a pharmaceutical compositions of the invention.
Another embodiment of the invention involves a method of treating or preventing a disease by administering to a subject in which such treatment or prevention is desired, a pharmaceutical composition in an amount sufficient to treat or prevent a disease. For example, the disease to be treated may include diabetes, colon cancer, respiratory ailments, neurodegenerative disorders, cardioplegia, and/or viral infections.
In another aspect, the invention involves a method of screening a phage library for transporter peptides, whereby a phage library is screened against specific cell types and ~it is then determined which cells have internalised phages.
In another embodiment, this method includes identifying the DNA of an internalised phage and deducing an expressed peptide from this.
In yet a further embodiment, this method includes a screening step whereby a phage library is panned for at least three cycles.
In still a fiuther embodiment, the invention includes a phage having a multivalent display of peptides.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows the structures for oximes 1 - 3. The branching Lys residues of the cores are acylated on both alpha and epsilon amino groups with the N-terminus of RRTK
peptide with or without a spacer. A reporter tag, FITC, together with a spacer is introduced into the core of each construct.
FIG. 2 shows the structures for oximes 4 - 6. The branching Lys residues of the cores are acylated on both alpha and epsilon amino groups with the N-terminus of RRTK
peptide together with a spacer. A reporter tag, biotin, together with a spacer is introduced into the core of each construct.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides peptide transporters and a peptide transport system that specifically targets various cell types for the intracellular delivery of drugs and therapeutic agents. Existing transport systems known in the art are often limited because they are inefficient, affect the host genome, alter the biological properties of the active substance (e.g., the effector), kill the target cell, or pose too high a risk to be used in a human subject (e.g., due to the use of viral conjugates). The peptide transport system of the present invention uses proprotein convertases and specific ligands thereof for the intracellular delivery of potential therapeutics, in order to overcome the limitations of transport systems known in the art. The present system exhibits efficient delivery of an unaltered biologically active substance that does not affect the host's genome and that is otherwise non-invasive.
Receptor-mediated endocytosis has been widely exploited in experimental systems for the targeted delivery of therapeutic agents into cells. Kato and Sugiyama, Crit. Rev. Ther. Drug Carrier. Syst. 14:287 (1997). Thus, cell-type specific delivery of peptides may be achieved by successfully targeting cell specific endocytotic receptors. Proprotein convertases are one example of a cell surface receptor which gets internalized through receptor mediated endocytosis. These proteins have been shown to be responsible for conversion of precursors of peptide hormones, neuropeptides, and many other proteins into their biologically active forms.
Cleavage sites for the proprotein convertase family contain the consensus R-X-X-R. Expression and localization information indicate that proprotein convertases transport extracellular ligands into the intracellular space.
For example, the mammalian proprotein convertases can be classified into three groups on the basis of their tissue distribution. In one class, Furin, PACE4, PCSlPC6, and LPCIPC7/PC8/SPC7 are expressed in a broad range of tissues and cell lines. In the second class, expression of the neuroendocrine-specific convertases PC2 and PC1/PC3 is limited to neuroendocrine tissues, such as pancreatic islets, pituitary, adrenal medulla and many brain areas. Further, in the third class, expression of PC4 is highly restricted to testicular spermatogenic cells. The neuroendocrine-specific convertases are mainly localized in secretory granules. PCS/PC6A has also been reported to be localized to secretory granules. Furthermore, indirect evidence has suggested that a proportion of proprotein convertases molecules is present on the cell surface, and it has been shown that furin cycles between the TGN
and the cell surface.
See Mandrup-Poulsen, BMJ. 316:1221 (1998).
Peptide ligands that mimic natural ligands are often identified using the phage display technique. The isolation of peptide sequences that direct efficient receptor-mediated endocytosis is enhanced by the use of phage display technologies. See Ivanenkov et al., Biochem. Biophys.
Acta 1448:450 and 463 (1999). Phage display libraries are extremely powerful tools that provide for a vast source of molecular variants including modifications of natural ligands to cell receptors (Cabibbo et al., Gene 167:41 (1995)) and short peptides (Zwick et al., Curr.
Opin. Biotechnol.
9:427 (1998)). Libraries have also been injected directly into mice where peptide sequences that show a 13-fold selectivity for brain and kidney have been successfully isolated. See Pasqualini and Ruoslahti, Nature 380:364 (1996); and Pasqualini and Ruoslahti, Mol.
Psychiatry 1:423 (1996).
Occasionally, the ligands obtained by this procedure may be of low affinity (micromolar range) due to the high degree of conformational freedom and small number of contact residues within a short peptide molecule. One way to improve the binding affinity of peptide ligands is to combine several copies of the low-affinity peptides in a single multimeric molecule. Avidities (and bioactivities) of the multimeric ligand can be greatly enhanced relative to that of a monomeric ligand. In some embodiments, multimeric constructs are generated from cell-targeting peptides as building blocks using oxime chemistry. The peptides can be derived, for example, from phage display experiments which select for peptide ligands which bind specifically to particular cells (e.g., pancreatic (3-cells) and are capable of eliciting uptake by these cells via receptor-mediated endocytosis.
For example, as a monomer, IgM is a low affinity protein, but, when in a pentameric form, its avidity is highly enhanced toward repetitive, antigenic determinants present on the surface of bacteria or viruses. See Roitt, Essential Immunology (OxfordBlackwell, London), pp.
65-84 (1991).
Similarly, a 105-fold enhancement occurred with the peptabody, a protein produced through recombinant DNA techniques. Alexey et al., Proc. Natl. Acad. Sci. USA
94:1663-1668 (1997). The low affinity of polypeptides derived from phage libraries is compensated by the pentameric structure of the peptabodies, resulting in high avidities towards their targets.
Recently, Rose also designed a similar a molecule named "chemobody". See Rose and Vizzavona, J. Am. Chem. Soc. 121:7034-7038 (1999). The chemobody is a synthetic molecule displaying multiple copies of a peptide subunit capable of binding non-covalently to a complementary structure, thus mimicking an antibody molecule. The amino acid sequence of .
the binding peptide was identified using phage library techniques and the subunits themselves o were assembled using oxime chemistry. In order to be able to bind a target through two or more peptide subunits simultaneously, an appropriate spacer and a linker were also introduced into the chemobody. Rose generated a model chemobody having four copies of a phage-derived peptide from influenza virus with flexible linkers.

Small transporter peptides that selectively target specific cell types may be derived from large phage display libraries, and used as monomeric or multimeric transporter molecules. The advantages of small peptide carriers such as those obtained using phage display libraries include high quality and purity, low immunogenicity, and the potential for highly efficient delivery to all cells in an organism Schwarze et al., Science 285:1573 (1999). Accordingly, peptide carriers have the potential to improve upon conventional transporters such as liposomes or viruses for the efficient delivery of a variety of macromolecules. See, for example, Mahato et al., J. Drug Target. 4:337 (1997); and Mahato et al., Crit. Rev. Ther. Drug Carrier. Syst.
14:133 4:337 (1997).
The transporter peptides described herein have a use in treating a variety of diseases, including, but not limited to, diabetes. In a normal organism, [3-cell mass is tightly regulated so that insulin secretion maintains normoglycemia. (i-cell mass is adjusted in response to the needs of the infant or adult organism, in particular as a reaction to certain physiological and physiopathological conditions. The appropriate (3-cell mass is essentially attained by a dynamic balance between (3-cell death and regeneration. This balance is achieved by differentiation of immature (3-cells and from the proliferation of preexisting insulin-secreting cells. See Scharfinann and Czernichow, Diabetes Metab. 22:223 (1996); and Bonner-Weir, Recent Prog.
Horm. Res. 49:91 (1994). In Type I diabetes, impaired balance of (3-cell mass results from accelerated (3-cell destruction, a process initiated by the specific attack of the immune system that targets pancreatic (3-cells. Preventing or decreasing the rate of (3-cell destruction may therefore not only help stabilize diabetes, but may also allow for islet regeneration to correct (3-cell mass insufficiency.
Several compounds have been established as potent tools used to decrease the rate of (i-cell loss in experimental models of Type I diabetes. Many of these molecules are peptidyl in nature, and thus are easily linked to peptide carriers. The peptides described herein serve as basis for the design of therapeutic "cargos", namely the coupling of the carriers (monomeric or multimeric "transporter peptides") with therapeutic agents ("effectors").
One embodiment of the transport system of the present invention targets (3-cell intracellular mechanisms for the treatment of Type I diabetes. Symptoms of Type I diabetes are secondary to the destruction of the pancreatic (3-cells by secretion of the immune system. See Mandrup-Poulsen, BMJ 316:1221 (1998). Conclusive data, both in human and rodents, indicates that the cytokine interleukin-1 (3 (IL-1 (3) in conjunction with TNFa and IFNy, secreted by macrophages and T-cells, are major components responsible for the final outcome that leads to (3-cell dysfunction and destruction and Type I diabetes. See Mandrup-Poulsen, Diabetologia 39:1005 (1996); Nerup et al., Diabetes Care 11 Suppl 1:16 (1988); and Mauricio and Mandrup-Poulsen, Diabetes 47:1537 (1998). These secreted cytokines engage in a highly complex network of signaling and effector molecules in pancreatic (3-cells. The signaling modifies the behavior of the cells and has a decisive impact on the cell fate. Accumulating evidence indicates that this regulatory intracellular network represents a promising taxget for the development of novel therapeutic approaches. See Iwahashi et al., Cytokines. Cell Mol. Ther.
4:45 (1998);
Sjoholm, Cell Death. Differ. 5:461 (1998); Stephens et al., J. Autoimmun.
10:293 (1997);
Rabinovitch et al., Diabetes 48:1223 (1999); Bleich et al., J. Clin. Invest.
103:1431 (1999);
Welsh et al., Mol. Med. 5:169 (1999); and Chen et al., Diabetes 49:562 (2000).
Each of the molecules involved in the treatment and integration of intracellular cytokine signaling may represent a target for transporter -drug design.
Among the most prominent signaling molecules recruited by IL-1 (3 in (3-cells are ceramides, prostaglandins, heat-shock proteins, the inducible NO synthase enzyme (iNOS), the transcription factor NF-xB (See Torgerson et al., J. Immunol. 161:6084 (1998)), and the three MAP kinases ERKI/2, p38 and JNK. Many of these molecules are targets for blockage with existing inhibitors that have led to improvement of [3-cell survival and function. iNOS knock our (KO) mice are resistant to IL-1(1 cytotoxicity (See Flodstrom et al., Diabetes 48:706 (1999)) and blockers of iNOS activity prevent different aspects of NO cytotoxicity (reviewed in Sjoholm, Cell Death. Differ. 5:461 (1998)). Islets and cell-lines studies have indicated that blockers of Ca2+ channels or caspase inhibitors prevent rodent (i-cell death. See Wang et al., Endocrinology 140:1200 (1999); Yamada et al., Diabetes 48:478 (1999). p38 inhibitors attenuate IL-1 (3-mediated inhibition of glucose-stimulated insulin release. See Larsen et al., J. Biol. Chem.
273:15294 (1998). [3-cell specific suppression of GAD expression in antisense GAD transgenic NOD mice prevented autoinimune diabetes. See Yoon et al., Science 284:1183 (1999).
Expression of bcl-2, IL-1 Ra as well as JBD (a dominant inhibitor of the c-Jun N-terminal Kinase JNK) in pancreatic (3-cell lines has lead to the generation of cells that resist apoptosis. See Bonny et al., Diabetes 50:77-82 (2000); Iwahashi et al., Diabetologia 39:530 (1996); Dupraz et al., Gene Ther. 6:1160 (1999); and Giannoukakis et al., Diabetes 48:1730 (1999). Together, these data indicate that the manipulation of intracellular events with specific tools holds great promise for the treatment of Type I diabetes.
One major challenge for disease treatment is to convert biologically important molecules into bioactive, cell-permeable compounds which are usable ira vivo. See Gibbs, Science 287:1969 (2000). For example, the most promising tools for the prevention of (3-cell loss are a number of large proteins (e.g., Bcl-2 (See Rabinovitch et al., Diabetes 48:1223 (1999)), inhibitors of cytokine signaling such as dominant negative versions of MyD88, TRAF, FADD or IRAK (See Burns et al., J. Biol. Chem. 273:12203 (1998); and Stephens et al., Endocrinology 140:3219 (1999)), or the JNK inhibitor JBD280 (See Ammendrup et al., Diabetes 49: 1468-1476 (2000)) that cannot be currently delivered ih vivo to tissues and cell-Types including pancreatic ~i-cells.
Recent work indicates progress in attempts to convert large proteins into small bioactive compounds which can be easily delivered to cells and organs. See Hawiger, Curr. Opin. Chem.
Biol. 3:89 (1999). These techniques essentially require two conditions: 1) a specific transporter or a chemical modification thereof is linked to the molecules for efficient delivery inside cells (see, for example, efficient short peptide transporters described in Schwaxze et al., Science 285:1573 (1999); Brugidou, et al., Biochem. Biophys. Res. Commun. 214:685 (1995); Oehlke et al., Biochim. Biophys. Acta 1414:127 (1998); and Terskikh et al., Proc. Natl.
Acad. Sci. U.S.A.
94:1663 (1997); and 2) the active portion of the protein has to be narrowed down so that small peptides sequence might be linked to the transporter. In short, these conditions generally define 3-30 amino acid long, bi-partite peptides that are able to enter cells while conserving the essential biological properties of the proteins from which they are derived.
As in cancer research (See Gibbs and Oliff, Cell 79:193 (1994)), there are numerous intracellular events in the [3-cells whose manipulations protect ~3-cells from cytokine-induced apoptosis --manipulations which appear to be promising targets for drug design.
Receptor-mediated endocytosis is widely exploited in experimental systems for the targeted delivery of therapeutic agents into cells. See Kato and Sugiyama, Crit. Rev. Ther. Drug Garner. Syst. 14:287 (1997). Endocytotic activity is a common property that has been described for many receptors including IgG Fc, somatostatin, insulin, IGF-I and II, transferrin, EGF, GLP-1, VLDL or integrin receptors. See Hofland et al., Proc. Assoc. Am.
Physicians. 111:63 (1999);
Anderson, Clin. hnmunol. hmnunopathol. 53:563 (1989); Lund et al., J. Biol.
Chem. 265:15713 (1990); Smith and Jarett, Lab. Invest. 58:613 (1988); Soler et al., Endocrinology 127:595 (1990);
Widmann et al., Biochem. J. 310:203 (1995); York et al., J. Biol. Chem.
274:1164 (1999); and Mukherjee et al., Physiol. Rev. 77:759 (1997). Cell-type specific receptors that mediate endocytosis have been reported. See Ivanenkov et al., Biochem. Biophys. Acta 1448:463 (1999).
Although strong experimental background indicates that transporter peptides which selectively target pancreatic (3-cells might be derived from large phage display libraries, no such attempts have been reported. The advantages of small peptide carriers such as those obtained using phage display libraries are numerous and include ease of generation by chemical synthesis, high quality and purity, low immunogenicity and potential for highly efficient delivery to all cells in an organism. See Schwarze et al., Science 285:1573 (1999).
Accordingly, the peptide carriers of the invention have the potential to perform better than conventional transporters, such as liposomes or viruses, in the efficient delivery of many macromolecules.
See, for example Mahato et al., J. Drug Target 4:337 (1997) and Mahato et al., Crit. Rev. Ther.
Drug Carrier. Syst.
14:133 (1997).
Phage peptide libraries are traditionally constructed in derivatives of the filamentous phage M13. Peptide libraries are fused to the minor coat protein pIII of the capsid that displays 1-5 copies of the peptide motif. See Zwick et al., Curr. Opin. Biotechnol.
9:427 (1998).
Alternatively, high-valent display is attained by using the major coat protein pVIII. However, these types of libraries have not been optimized for the isolation of receptor-mediated endocytotic peptide sequences. The following considerations are relevant for the recovery of carriers with the highest efficiencies of internalization: 1) mono- or low-valent display of peptides is essentially insufficient for the efficient uptake of such large structures as filamentous phages, however multivalent display allows for efficient uptake (See Ivanenkov et al., Biochem.
Biophys. Acta 1448:450 (1999)); and 2) the internalization of receptor-bound ligands involves concentration of cell surface receptors in specialized areas of the plasma membrane and subsequent formation of clathrin-coated vesicles (See Damke, FEBS Lett. 389:48 (1996)).
The large size of the M13 derivatives (1-1.5 Nxn) (See Zacher et al., Gene 9:127 (1980)) exceeds the typical size of classical clathrin-coated pits (150 nM). Clathrin-coated pits are invaginated structures on the plasma membrane that occupy approximately 2% of the membrane surface. These specialized structures direct the highly efficient receptor-mediated internalization process that clears extracellular proteins or peptides such as insulin or EGF
at the extremely rapid rate of 10-50%/min. See Mukherjee et al., Physiol. Rev. 77:759 (1997).
Thus, receptor-mediated internalization by these specialized and highly efficient structures is not expected to occur with the conventional M13 phages.
Accordingly, published attempts have failed to produce peptides that display a high internalization rate of peptide bearing phages. To date, no consensus internalization motif specific for a particular cell-type has emerged from these studies.
In certain aspects, the invention described herein relates to the identification of transporter peptides which promote the internalization of peptide-bearing effectors. Once the peptide sequences are determined, the peptides are bound to effector molecules in order to transport the effector molecules across a biological membrane. For example, a series of peptides that target pancreatic insulin secreting j3 cells were isolated by phage display panning.
Monomeric and multimeric constructs employing these cell-targeting peptides as building blocks were synthesized. These constructs represent novel chemical entities designed to specifically deliver therapeutic agents to control diabetes.
Transport peptides of the invention can be represented by Formula I:
[P]~ [Spacer]X Core-[Linker]y-[Reporter]Z
where P is a peptide, falling within the consensus motifs (XmRXoRXn); (Xm~n)i (XmRR~~tX"); and (XmR~~RXn), as described above. For example, the peptide can include the sequence Xl-X~_X3-X4, where Xl and X4 can be R or I~ and Xi and X4 can be any amino acid. In one embodiment, the invention provides a multimeric transporter with at least one peptide having the amino acid sequence R-X-X-R. In other embodiments, the invention provides a multimeric transporter peptide with at least one peptide having an amino acid sequence of any one of SEQ
ID NOS: 1-34. In various other embodiments, the transporter peptides are derived from protein convertase ligands. In still other embodiments, the transporter peptides are derived from protein convertase cleavage sites. The number of peptides in a given transporter, represented by v, is an integer. In monomeric transporter, v is one. In a multimeric transporter, v is an integer from two to eight or more. In any given multimeric conjugate, each of the peptides may be the same as or different from each other.
A spacer may be present or absent between the peptide and core, thus x is one or zero.
Preferred spacers are flexible, amphiphilic, non-immunogeilic and unsusceptible to proteases, and include polyethylene glycols (PEG) such as succinimidyl-PEG (succ-peg).
As used herein, a "core" is a structure that serves to connect the peptides within a multimeric transporter peptide. The core also serves as an attachment point for linker, spacer, and reporter groups. Typically, a core will have many functionalities available for attachment so that the various different moieties (the peptides and spacers, linkers, and reporter, if present) of the multimeric construct can be fused to the core, thereby generating one molecule. The core moieties contemplated include the polylysine (K) cores described in detail below and also include those structures having a built-in linker. The chosen core determines the number of peptides present within a given conjugate. Typical cores may include: C4-KaK-K(succ-peg-S)-amide; C-GGG -[K(C)]3-K(succ-peg-S)-amide; (NH20CH2C0)4-KZK-K(succ-peg)-amide;
(NHaOCH2C0)$-K4KZK-K(GGG)-amide. In a monomeric transporter peptide, the core may be present or absent.
In the transporter construct, the linker and reporter groups can be present or absent, thus y and z are, independently, one or zero in Formula I.
As used herein, a "transporter peptide" is a peptide that facilitates the translocation of a substance across a biological membrane. Unless otherwise specified, or inaccurate in the context used, reference to a transporter peptide encompasses a monomeric or multimeric transporter peptide.
A transporter peptide is a peptide that facilitates the passage, or translocation, of a substance across a biological membrane, particularly into the cytoplasm or nucleus, of the cell.
Translocation may be detected by various procedures, including a cellular penetration assay as described in, for example, PCT application No. WO 97/02840. Generally, a cellular penetration assay is performed by: a) incubating a cell culture with a translocating peptide; b) fixing and permeabilizing the cells; and c) detection of the presence of the peptides inside the cell. The detection step may be carried out by incubating the fixed, permeabilized cells with labeled antibodies directed to the peptide, followed by detection of an immunological reaction between the peptide and the labeled antibody. Alternatively, detection may also be achieved by using a detectably labeled peptide, and directly detecting the presence of the label in cellular compartments. The label may be, for example, a radioactive label, or a fluorescent label, a dye, or any label capable of detection. See, e.g., de Jong et al., J. Nucl. Med.
40:2081 (1999); and Breeman et al., Int. J. Cancer 81:658 (1999).
The invention further includes transport units, or conjugates, which are complexes of the transporter peptide coupled or fused to an effector. As used herein, the term "coupled" means any type of interaction enabling a physical association between an effector and the peptide. The association may be covalent or a non-covalent in nature, and it must be sufficiently strong so that the vector does not disassociate before or during translocation. Coupling may be achieved using any chemical, biochemical, enzymatic or genetic coupling known to those skilled in the art. The effector of interest may be coupled to the N-terminal or C-terminal end of the peptide vector.
As used herein, the terms "fused" or "fusion" or "associates" or "interacts"
are meant to include all specific interactions that result in two or more molecules showing a preference for one another relative to some third molecule, including processes such as covalent, ionic, hydrophobic, and hydrogen bonding, but does not include non-specific associations such as solvent preferences.
In some embodiments, the transporter peptide is coupled to or fused to one or more effectors. The coupling can be direct or can be through a core moiety. The "effector" can be any suitable molecule, including DNA, RNA, a protein, a peptide, or a pharmaceutically active agent, such as, for example, a toxin, an antibiotic, an antipathogenic agent, an antigen, an antibody, an antibody fragment, an ixnmunomodulator, an enzyme, or a therapeutic agent.
When more than one effector is present, the effectors can be the same or different. An effector refers to any molecule or compound of, for example, biological, pharmaceutical, diagnosis, tracing, or food processing interest. Effectors may consist of nucleic acids (ribonucleic acid, deoxyribonucleic acid) from various origins, and particularly of human, viral, animal, eukaryotic or prokaryotic, plant, synthetic origin, etc. A nucleic acid of interest may be of a variety of sizes, ranging from, for example, a simple trace nucleotide to a genome fragment, or an entire genome. Likewise, it may be a viral genome or a plasmid. Alternatively, the effector of interest may also be a protein, such as, for example, an enzyme, a hormone, a cytokine, an apolipoprotein, a growth factor, an antigen, or an antibody, etc. Furthermore, the effector may be a pharmaceutically active agent, such as, for example, a toxin, a therapeutic agent, or an antipathogenic agent, such as an antibiotic, an antiviral, an antifimgal, or an anti-parasitic agent. The effector of interest may itself be directly active or may be activated ih situ by the peptide, by a distinct substance, or by environmental conditions.
The transporter peptide conjugates of the invention can be represented by Formula (IIJ:
([P]~ [Spacer]X Core-[Linker]y-[Reporter]Z)~E
where P, Core, Spacer, Linker, Reporter, v, x, y, and z are as described above and E is an effector. In a covalently fused conjugate, the effector can be attached to the core, to the linker, or to both, as the laws of chemistry apply. In a monomeric transporter peptide conjugate (v =1), the core can be present or absent.
The term "pharmaceutically active agent" is used herein to refer to a chemical material or compound which, when administered to a human or animal organism induces a detectable pharmacologic and/or physiologic effect. The term "therapeutic agent" is used herein to refer to a chemical material or compound which, when administered to a human or animal organism induces a desired pharmacologic and/or physiologic effect.
The transporter peptides according to the present invention are characterized by the fact that their penetration capacity is virtually independent of the nature of the substance of the interest (the effector) that is coupled to it.
The invention also includes a method of introducing an substance of interest into a cell or a cell nucleus. The method includes contacting the cell with a transporter peptide-effector conjugate or transporter unit in an amount sufficient to enable efficient penetration into the cells.
In general, the method may be used for in vivo or ih vitro internalization of the conjugate. For example, the conjugate can be provided in vitro, ex vivo, or in vivo.
Furthermore, a transporter peptide according to this invention may be capable of potentializing the biological activity of the coupled substance. Therefore, a transporter peptide can increase the biological activity of the effector to which it is coupled. In an in vitro method, an effector is coupled to a transporter, and the conjugate is incubated with cells at a temperature which enables active metabolism of the cells. In some cases, the transporter-effector conjugate is injected into particular cells. Those skilled in the art will recognize that any other method of introducing the conjugate into the cells can also be used.
In addition to the peptide-effector conjugates, the invention also provides a pharmaceutically acceptable base or acid addition salt, hydrate, ester, solvate, prodrug, metabolite, stereoisomer, or mixture thereof. The invention also includes pharmaceutical formulations comprising a peptide-effector conjugate in association with a pharmaceutically acceptable carrier, diluent, or excipient.
Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention which are generally prepared by reacting the free base with a suitable organic or inorganic acid to produce "pharmaceutically-acceptable acid addition salts" of the compounds described herein. These compounds retain the biological effectiveness and properties of the free bases. Representative of such salts are the water-soluble and water-insoluble salts, such as the acetate, amsonate (4,4-diaminostilbene-2, 2'-disulfonate), benzenesulfonate, benzonate, bicarbonate, bisulfate, bitarirate, borate, bromide, butyrate, calcium edetate, camsylate, carbonate, chloride, citrate, clavulariate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexafluorophosphate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, N-methylglucamine ammonium salt, 3-hydroxy-2-naphthoate, oleate, oxalate, palmitate, pamoate (1,1-methylene-bis-2-hydroxy-3-naphthoate, embonate), pantothenate, phosphate/diphosphate, picrate, polygalacturonate, propionate, p-toluenesulfonate, salicylate, stearate, subacetate, succinate, sulfate, sulfosaliculate, summate, tannate, tartrate, teoclate, tosylate, triethiodide, and valerate salts.
According to the methods of the invention, a human patient can be treated with a pharmacologically effective amount of a peptide or conjugate. The term "pharmacologically effective amount" means that amount of a drug or pharmaceutical agent (the effector) that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by a researcher or clinician.
The invention also includes pharmaceutical compositions suitable for introducing an effector of interest into a cell or cell nucleus. The compositions are preferably suitable for internal use and include an effective amount of a pharmacologically active compound of the invention, alone or in combination, with one or more pharmaceutically acceptable Garners. The compounds are especially useful in that they have very low, if any, toxicity.
Preferred pharmaceutical compositions are tablets and gelatin capsules comprising the active ingredient together with a) diluents, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone; if desired d) disintegrants, e.g., starches, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or e) absorbents, colorants, flavors and sweeteners. Injectable compositions are preferably aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions. The compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure andlor buffers.
In addition, they may also contain other therapeutically valuable substances. The compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1 to 75%, preferably about 1 to 50%, of the active ingredient.
Administration of the active compounds and salts described herein can be via any of the accepted modes of administration for therapeutic agents. These methods include systemic or local administration such as oral, nasal, parenteral, transdermal, subcutaneous, or topical administration modes.
Depending on the intended mode of administration, the compositions may be in solid, semi-solid or liquid dosage form, such as, for example, injectables, tablets, suppositories, pills, time-release capsules, powders, liquids, suspensions, or the like, preferably in unit dosages. The compositions will include an effective amount of active compound or the pharmaceutically acceptable salt thereof, and in addition, and may also include any conventional pharmaceutical excipients and other medicinal or pharmaceutical drugs or agents, carriers, adjuvants, diluents, etc., as are customarily used in the pharmaceutical sciences.
For solid compositions, excipients include pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like may be used. The active compound defined above, may be also formulated as suppositories using for example, polyalkylene glycols, for example, propylene glycol, as the carrier.
Liquid, particularly injectable compositions can, for example, be prepared by dissolving, dispersing, etc. The active compound is dissolved in or mixed with a pharmaceutically pure solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form the injectable solution or suspension.
If desired, the pharmaceutical composition to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, and other substances such as for example, sodium acetate, triethanolamine oleate, etc.
Parental injectable administration is generally used for subcutaneous, intramuscular or intravenous injections and infusions. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions or solid forms suitable for dissolving in liquid prior to injection.

One approach for parenteral administration employs the implantation of a slow-release or sustained-released systems, which assures that a constant level of dosage is maintained, according to U.S. Pat. No. 3,710,795, incorporated herein by reference.
The compounds of the present invention can be administered in such oral dosage forms as tablets, capsules (each including timed release and sustained release formulations), pills, powders, granules, elixers, tinctures, suspensions, syrups and emulsions.
Likewise, they may also be administered in intravenous (both bolus and infusion), intraperitoneal, subcutaneous or intramuscular form, all using forms well known to those of ordinary skill in the pharmaceutical arts.
The dosage regimen utilizing the compounds is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound or salt thereof employed. An ordinarily skilled physician or veterinarian can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
Oral dosages of the present invention, when used for the indicated effects, may be provided in the form of scored tablets containing 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100.0, 250.0, 500.0 or 1000.0 mg of active ingredient.
Compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three or four times daily.
Furthermore, preferred compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art.
To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen. Other preferred topical preparations include creams, ointments, lotions, aerosol sprays and gels, wherein the concentration of active ingredient would range from 0.1 % to 15%, w/w or w/v.
The compounds herein described in detail can form the active ingredient, and are typically administered in admixture with suitable pharmaceutical diluents, excipients or carriers (collectively referred to herein as "carrier" materials) suitably selected with respect to the intended form of administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with conventional pharmaceutical practices.

For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
The compounds of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellax vesicles. Liposomes can be formed from a variety of phospholipids, containing cholesterol, stearylamine or phosphatidylcholines. In some embodiments, a film of lipid components is hydrated with an aqueous solution of drug to a form lipid layer encapsulating the drug, as described in U.S. Pat. No. 5,262,564.
The compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropyl-methacrylamide-phenol, polyhydroxyethylaspanamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues. Furthermore, the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
Any of the above pharmaceutical compositions may contain 0.1-99%, preferably 1-70%
of the active compounds, especially compounds of the Formula I as active ingredients.
OTHER EMBODIMENTS
The details of one or more embodiments of the invention have been set forth in the accompanying description above. From the foregoing detailed description of the specific embodiments of the invention, it should be apparent that unique methods and compositions for translocation across a biological membrane have been described. Although particular embodiments have been disclosed herein in detail, this has been done by way of example for purposes of illustration only, and is not intended to be limiting with respect to the scope of the appended claims that follow. In particular, it is contemplated by the inventor that various substitutions, alterations, and modifications may be made to the invention without departing from the spirit and scope of the invention as defined by the claims. For instance, the choice of the particular type of cell, or the particular effector to be translocated is believed to be a matter of routine for a person of ordinary skill in the art with knowledge of the embodiments described herein.
In the specification and the appended claims, the singular forms include plural referents unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications cited in this specification are incorporated by reference.
The following EXAMPLES are presented in order to more fixlly illustrate the preferred embodiments of the invention. These EXAMPLES should in no way be construed as limiting the scope of the invention, as defined by the appended claims.
EXAMPLES
Example I: Identification of internalization peptide motifs.
Phage peptide libraries are traditionally constructed in derivatives of the filamentous phage M13. Peptide libraries are fused to the minor coat protein pIII of the capsid that displays 1-5 copies of the peptide motif. See Zwick et al., Curr. Opin. Biotechnol.
9:427-436 (1998).
Alternatively, high valent display is attained by using the major coat protein pVIII. These libraries however have not been optimized for the isolation of receptor-mediated endocytotic peptide sequences, because mono- or low-valent display of peptides is insufficient for efficient uptake of such giant structures as filamentous phages. Only a multivalent display allows for efficient uptake. See Ivanenkov et al., Biochim. Biophys. Acta 1448:450-462 (1999). In addition, the internalization of receptor-bound ligands involves concentration of cell surface receptors in specialized areas of the plasma membrane and subsequent formation of clathrin-coated vesicles (See Damke FEBS Lett. 389:48-51 (1996)), and receptor-mediated internalization by these specialized and highly efficient structures is not expected to occur with the conventional M13 phages.
Phage display libraries in the T7 415 phage system and in the T7 413b phage systems were constructed. This phage exhibits 415 copies of the displayed peptides within a reduced volume (capsid is ~SOnM diameter). Therefore, included in this invention is a phage display library in a novel phage system that fulfills the following criteria:
multivalent display of 4-50-mer peptides (>400 copies/phage); small size (50 nM); efficient recovery of internalized phages;
removal of non-internalized bound-phages; and large number of individual peptide sequences (3x10$ independent clones, representing >109 heptapeptide sequences).
This library has been successfully used for the isolation of a peptide motif that directs efficient and specific intracellular delivery of macromolecules to the (3TC-3 cell-model. In addition, this library has been used with five different (non-~i-) cell lines, and in each case, the enrichment of peptide motifs specific for each cell-type has been observed. A
general overview of the procedure is outlined below.
Selectiorzlenrichment procedure A phage display library is panned against a number of insulin-secreting cell-lines, rodent and human isolated islets and FACS purified (3-cells, and finally is injected directly into animals (mice, rats, pigs) before extracting islets and recovering internalized phages. The panning procedure consists of at least three cycles of phage addition, recovery and amplification.
Alternatively, and in order to isolate the most selective ligands, phages that bind other cell-types are subtracted by incubating the library with different non-insulin secreting cells before panning the library against (3-cells. Experiments involving chloroquine to block lysosomal degradation.
are performed as described. See Ivanenkov et al., Biochem. Biophys. Acta 1448:450 (1999).
Determination ofphage specificity Panned phages are isolated and incubated with a number of different cells and organs.
For example, in certain experiments, panned phages are incubated with insulin and non-insulin secreting cells and organs. Uptake is determined by counting the number of phages recovered.
Immunocytochemistry studies are performed with anti-phage antibodies.

Characterization of phage-beard peptides DNA from isolated phages are sequenced and expressed peptides are deduced.
Peptides which direct internalization and mutated versions of these peptides are chemically synthesized and N-terminally labeled with FITC or iodinated. Labeled peptides are added to different cell types, isolated rodent and human islets, and directly injected into mice.
Specificity of uptake, subcellular localization, clearance and stability are estimated. See Widmann et al., Biochem. J.
310:203 (1995).
Biochemical assays For analysis of insulin and non-insulin secreting cells, characterized peptides are linked to three known sequences: YVAD (caspases inhibitor, SEQ ID N0:35; Rouquet et al., Curr.
Biol. 6:1192 (1996)), VQRKRQKLMP (inhibitor of NF-xB nuclear localization (Lin et al., J.
Biol. Chem. 270:14255 (1995), SEQ ID N0:36) or RPKRPTTLNLFPQVPRSQDT (JNK
inhibitor, Bonny et al., Diabetes 50:77-82 (2000), SEQ ID N0:37). These peptides are chemically synthesized and added to insulin and non-insulin secreting cells.
Caspases, NF-xB
and JNK are activated by the general activator etoposide (See Kim et al., Anticancer Res. 20:439 (2000)) or anisomycin (See Usami et al., Biochem. Pharmacol. 55:185 (1998)).
Inhibition of caspases, NF-xB and JNK by the peptides are studied in [3- and non [3-cells.
These experiments indicate whether the peptide carriers transport potential drugs in an active conformation specifically inside the /3-cells.
Uptake ofpotential tlaerapeutic agents by the GLP-1 receptor Expression of the GLP-1 receptor (GLP-1R) is mainly restricted to the brain and the pancreas. See Yamato et al., Horm. IVIetab. Res. 29:56 (1997). The receptor is internalized following binding to an agonist. See Widmann et al., Biochem. J. 310:203 (1995). These properties make the GLP-1R an attractive tool to mediate preferential delivery of therapeutic agents to the pancreatic (3-cells. This property is evaluated as described above. Information gathered with the GLP-1R assists, for example, in the design of bispecific dimers with enhanced selectivity.

Identification of othef° internalization motifs for the GLP-1 receptor COS-7 cells transfected with the GLP-1R serve as substrates for a panning experiments as above. Newly identified motifs are evaluated for their specificity and their capacity to direct endocytosis.
Production of all D-retro-inverso peptides In some embodiments, the peptides can be synthesized as retro-inverso peptides. All-D-retro-inverso peptides with increased stability and lower immunogenicity (See Sela and Zisman, FASEB J. 11:449 (1997)) are analyzed as described above.
Evolution has ensured the almost exclusive occurrence of L-amino acids in naturally occurring proteins. Virtually all proteases therefore cleave peptide bonds between adjacent L-amino acids; thus, artificial proteins or peptides composed of D-amino acids are largely resistant to proteolytic breakdown. This resistance has been attractive to drug designers, but the exclusivity of biological systems for proteins made of L-amino acids means that such proteins cannot interact with the mirror image surface formed by enantiomeric proteins.
Thus, an all D-amino acid protein usually has no biological effect or activity.
Linear modified retro-peptide structures have been studied for a long time (See Goodman et al., Accounts of Chemical Research, 12:1-7 (1979)) and the term "retro-isomer" was designated to include an isomer in which the direction of the sequence is reversed compared with the parent peptide. By "retro-inverso isomer" is meant an isomer of a linear peptide in which the direction of the sequence is reversed and the chirality of each amino acid residue is inverted;
thus, there can be no end-group complementarity.
More recently, Jameson et al. engineered an analogue of the hairpin loop of the CD4 receptor by combining these two properties: reverse synthesis and a change in chirality. See Jameson et al., Nature 368:744-746 (1994) and Brady et al., Nature, 368:692-693 (1994). The net result of combining D-enantiomers and reverse synthesis is that the positions of carbonyl and amino groups in each amide bond are exchanged, while the position of the side-chain groups at each alpha carbon is preserved. Jameson et al. demonstrated an increase in biological activity for their reverse D peptide, which contrasts to the limited activity in vivo of its conventional all-L enantiomer (due to its susceptibility to proteolysis).
A partially modified retro-inverso pseudopeptide has been reported for use as a non-natural ligand for the human class I histocompatibility molecule, HLA-A2.
See Guichard et al., Med. Chem. 39:2030-2039 (1996). Such non-natural ligands had increased stability and high MHC-binding capacity.
Retro-inverso peptides are prepared for peptides of known sequence in the following manner. A peptide having a known sequence (e.g., a tumor antigen peptide) is selected as a model peptide for designing and synthesizing a retro-inverso peptide analog.
The analog is synthesized using D-amino acids by attaching the amino acids in a peptide chain such that the sequence of amino acids in the retro-inverso peptide analog is exactly opposite of that in the selected peptide which serves as the model. To illustrate, if the peptide model is a peptide formed of L-amino acids having the sequence ABC, the retro-inverso peptide analog formed of D-amino acids would have the sequence CBA. The procedures for synthesizing a chain of D-amino acids to form the retro-inverso peptides are known in the art and are illustrated in the above-noted references.
Since an inherent problem with native peptides is degradation by natural proteases, the peptides of the invention may be prepared to include the "retro-inverso isomer" of the desired peptide. Protecting the peptide from natural proteolysis should therefore increase the effectiveness of the specific heterobivalent or heteromultivalent compound.
A higher biological activity is predicted for the retro-inverso containing peptide when compared to the non-retro-inverso containing analog owing to protection from degradation by native proteinases.
Production of modified peptides In some embodiments, the peptides can be synthesized as modified peptides. The modified peptides are be analyzed as described above.
Analogs can differ from the native peptides by amino acid sequence, or by modifications which do not affect the sequence, or by both. Preferred analogs include peptides whose sequences differ from the wild-type sequence (i.e., the sequence of the homologous portion of the naturally occurring peptide) only by conservative amino acid substitutions, preferably by only one, two, or three, substitutions, for example, substitution of one amino acid for another with similar characteristics (e.g., valine for glycine, arginine for lysine, etc.) or by one or more non-conservative amino acid substitutions, deletions, or insertions which do not abolish the peptide's biological activity.

Modifications (which do not normally alter primary sequence) include izz vivo or izz vitro chemical derivitization of peptides, e.g., acetylation or carboxylation. Also included are modifications of glycosylation, e.g., those made by modifying the glycosylation patterns of a peptide during its synthesis and processing or in further processing steps, e.g., by exposing the peptide to enzymes which affect glycosylation e.g., mammalian glycosylating or deglycosylating enzymes. Also included are sequences which have phosphorylated amino acid residues, e.g., phosphotyrosine, phosphoserine, or phosphothreonine.
The invention includes analogs in which one or more peptide bonds have been replaced with an alternative type of covalent bond (a "peptide mimetic") which is not susceptible to cleavage by peptidases. Where proteolytic degradation of the peptides following injection into the subject is a problem, replacement of a particularly sensitive peptide bond with a noncleavable peptide mimetic will make the resulting peptide more stable and thus more useful as a therapeutic. Such mimetics, and methods of incorporating them into peptides, are well known in the art. Also useful are amino-terminal blocking groups such as t-butyloxycarbonyl, acetyl, theyl, succinyl, methoxysuccinyl, suberyl, adipyl, azelayl, dansyl, benzyloxycarbonyl, fluorenylinethoxycarbonyl, methoxyazelayl, methoxyadipyl, methoxysuberyl, and 2,4,-dinitrophenyl. Blocking the charged amino- and carboxy-termini of the peptides would have the additional benefit of enhancing passage of the peptide through the hydrophobic cellular membrane and into the cell.
Production of multimeric peptides Multimeric ligands display enhanced avidity of up to several orders of magnitude that translates in enhanced rate of internalization. See Terskikh et al., Proc.
Natl. Acad. Sci. LT.S.A.
94:1663 (1997); York et al., J. Biol. Chem. 274:1164 (1999). Monospecific dimers display great avidity and bispecific dimers are likely to have greater selectivity that may enhance their practical potential as specific cell-targeting agents. See Caruthers and Lerner, Chem. Biol. 3:537 (1996). Monomeric and multimeric peptides (both mono-and pluri-specific) can be synthesized as peptides or as peptidomimmetics, e.g., with either flexible peptidyl or sugar-based backbones.
See Caruthers and Lerner, Chem. Biol. 3:537 (1996); Zeng et al., J. Pept. Sci.
2:66 (1996);
LTlbrich et al., J. Controlled Release 64.(1.-3.):63.-79. 64:63 (2000).

Intracellular localization The different peptide sequences isolated may localize to different cell compartments (e.g., the nucleus, mitochondria, cytosol, etc.). This is evaluated using labeled (e.g., iodinated or FITC-labeled) peptides. This localization information is used for the design of the functional studies. For example, peptides accumulating in the cytosol are preferred for inhibiting NF-xB
nuclear translocation, while peptides entering the nucleus are best suited for inhibiting JNK. In some embodiments, sequences such as nuclear localization motifs can be added to a conjugate to redirect the Garners to the appropriate cellular compartment.
Functional studies (3-cell targeting transporter peptides (e.g., L- or D- enantiomers, monomeric or multimeric) linked to the caspase, NF-oB or JNK inhibitors are added to ~3-cell lines, FACS
purified (3-cells and isolated human and rodent islets. Apoptosis is induced by IL-(3 (in conjunction with TNFa and IFN~y) and resistance to apoptosis is evaluated.

In vivo diabetes experimerats NOD mice are injected in pre- and post-diabetic states with the effector peptides ((3-cell targeting peptides linked to the caspase, NF-xB or JNK inhibitors). Dose and frequency of injection is determined as described above. Occurrence of diabetes is then measured.
Imnaunogenicity assays The immunogenic potential of the peptides is evaluated in rodents and rabbits.
Cloning Peptide motifs that direct efficient uptake by specific cells are described in Examples III
and IV. These peptides are used for the cloning and characterization of the cognate receptors from e.g., INS-1, (3TC-3 and human islet cDNA libraries using established procedures. See Thorens, Proc. Natl. Acad. Sci. U.S.A. 89:8641 (1992); Volz et al., FEBS Lett.
373:23 (1995).
Characterization Tissue distribution of the cloned receptors) is evaluated by Northern and Western blotting of insulin and non-insulin secreting cells and organs. Binding kinetics, clearance and specificity of uptake are evaluated by transient transfection of the receptors in COS-7 cells.

Control peptides are mutated sequences as well as known peptides such as, for example, GLP-1, GIP, glucagon, secretin, etc. Alternative internalization motifs for these receptors are characterized by panning the library in transfected COS-7 cells as described above.
Example II: Methods of screening for transporter peptides.
Unless otherwise specified, all solvents and reagents were obtained from Fluka, Buchs, Switzerland, were of analytical or higher grade and were used without further purification. All amino acids were purchased from Peptide Institute Inc.(Japan). Resins were from Applied Biosystems, USA; Novabiochem, Switzerland, or Bachem, Switzerland. Water was repurified using a Milli-Q system (Millipore, Inc.). The insulin-secreting cell line (3TC-3 was used in the bioassay. See Efrat et al., Proc. Natl. Acad. Sci. USA 85:9037-9041.
Phage preparation and enrichment procedures A library of 3x10$ independent phages displaying random 15-mer epitopes at the surface of the capsids was generated using standard procedures (e.g., in T7 415 or the T7 413b phage, Novagen). See Smith and Scott, Methods Enzymol. 217:228 (1993). Phages were amplified then purified by polyethylene glycol (PEG) precipitation and finally resuspended at a concentration of 101° infective particles per ~,l in Tris-EDTA buffer (10:1 mM, TE) as described (by Smith and Scott, supra). Phages (1012) were added to cells in culture medium for 1 to 24 hours. Longer incubation times were preferred to favor isolation of phages that escaped proteolytic degradation in endocytotic vesicles. Following binding and internalization, cells were washed and non-internalized phages were destroyed by digestion with subtilisin (3 mg/ml) (44). Following extensive washing, internalized phages were then recovered by lysing cells in a buffer containing 2% deoxycholate, 10 mM Tris-HCl and 2mM EDTA, pH 8Ø
Recovered phages were finally amplified in E. coli cells (XI,-1-Blue) and purified as described above. This preparation of selected phages was then used for a second round of panning.
Three to five sequential rounds were performed to obtain enrichment of specific phage-bearing peptide sequences.

Immunocytochenaistry and fluorescence studies Single phages isolated according to the enrichment scheme above were amplified and added for 24 hours to cells in culture medium. Medium was then washed off and cells fixed in cold methanol-acetone (1:1) for 5 minutes. Antibodies directed against the phage capsid were used with a fluorescein-conjugated secondary antibody. Classical fluorescence microscopic studies and confocal microscopic assays were performed. Tissues were embedded in paraffin before processing.
Peptides Peptides were synthesized using classical F-moc chemistry (Auspep, Australia), with a C-terminal amide group and labeled with FITC or iodinated when necessary. All peptides were purified by HPLC and analyzed by Mass-Spectrometry.
Biochemical studies Peptides are added one hour before JNI~, NF-xB and caspases are activated in different cell-lines, such as, for example, (3TC-3, INS-l, HeLa, WiDr, HepG2, NIH 3T3, COS-7, by etoposide (VP-16, Alexis) for 1 hour. Cell extracts are processed for JNK
activity (using a solid phase JNK assay with c-Jun as substrate (See Bonny et al. J. Biol. Chem.
275:16466 (2000)), NF-xB nuclear translocation (electrophoretic mobility shift assay (See Lin et al., J. Biol. Chem.
270:14255 (1995)) and caspase activity (using available commercial kits and antibodies, Upstate Biochemicals).
Measurement of apoptosis Apoptosis is measured using a combination of Hoechst 33342 and propidium iodide as previously described. See Bonny et al. J. Biol. Chem. 275:16466 (2000);
Hoorens et al., J. Clin.
Invest. 98:1568 (1996).
Islets Islets are isolated by the method of Gotoh et al. (Gotoh et al., Transplantation 43:725 (1987)). Human islets can be obtained, e.g., from the "Insel Spital", Bern, Switzerland.

lllice The exact dosage and time-frame of injection is optimized for each peptide.
However, previous experience with JNI~I peptides indicates that 100 wl of a 1 mM
solution of peptides in PBS injected every two days is a reasonable starting point.
,ZAP-Expf~ess library The INS-1 cDNA library in the ,ZAP-Express prokaryotic/eukaryotic expression vector has been used to clone the IB1 and IB2 cDNAs. Bonny et al., J. Biol. Chem.
273:1843 (1998);
Negri et al., Genomics 64:324 (2000). This library is easily converted to a plasmid library under the control of the eukaryotic CMV promoter by simple helper phage excision (Stratagene).
Example III' Pannin~of~ha a dis la~librarX and characterization of internalized peptide motifs The ability of specifically targeting (3-cells for drug delivery will have an enormous 1 S impact on the treatment of Type I diabetes. Blockers of (3-cell destruction that do not essentially alter [3-cell function (i.e. insulin secretion) already exist (e.g. JBD, bcl-2). The conversion of one of these molecules (JBD) into a small peptide has been shown to retain full biological activity.
The pancreatic [3-cell line [3TC-3 was panned with the phage display library mentioned herein. Selective enrichment of the number of recovered phages was observed at each cycle of selection as seen below in Table 1. Panning experiments using (3TC-3 cells were performed with 109 phages used at each step of the enrichment procedure. The number of phages recovered at 0 °C (no endocytosis) is less than 100, indicating that the background of extracellular, but not internalized, bound phages is extremely low under the conditions described herein.
Table 1.
PanningPha es recovered 1St <1x10 2" 2x10 3r 3x10 The occurrence of phages recovered after three steps of panning in the [3TC-3 cell line is seen in Table 2.

Table 2.
P1 61%

P6 17%

P8 5.5%

P10 5.5%

P65 5.5%

P66 5.5%

Titration experiments were performed with the phage P1 (SEQ ID NO:1) incubated with [3TC-3 cells for the times indicated in Table 3. The ratio of input/recovered phages is also shown. Titration experiments indicated that as much as 10% of the initial P1 phage input could be recovered.
Table 3.
Phage Incubation% Recovered P1 lh 0.01 Sh 1 17h 10 Determination of the specificity of uptake was performed by titrating the number of recovered phages in 5 different cell lines. Phages (10g) were incubated for 16 hours with the indicated cell lines, and the number of internalized and recovered phages was calculated as seen in Table 4. Control phages displaying an integrin internalization motif showed a similar number (1-3x106) of recovered phages for all cell lines. This indicates that P1 (SEQ
ID NO:1, see Table 1 S 5) is taken up by (3TC-3 cells 10,000 to 1,000,000 fold more efficiently than by any other cell line tested.
Table 4.
Cells Pha es recovered TC-3 1x10 HeLa <1x10 WiDr 2x10 He <10 Peptides were then synthesized from the sequence of the displayed peptide in phage P1.
The sequence of the P1 5-mer peptide was linked to a 10 amino-acid random sequence that was labeled with FITC. The control sequence was identical except that the P1 5-mer sequence was replaced by (Ala)5. Peptides (10 pM) were added to the cells for one hour and cells were washed and fixed in cold methanol-acetone (1:1). FITC-labeled P1 peptides could be visualized inside (3TC-3 cells, but not in other cell types.
Sequence analysis of 20 recovered phages at the final cycle of enrichment is shown in Table 5. Importantly, all sequences strictly obeyed to a same conserved consensus sequence of 5 amino acids. This suggests the specific selection/enrichment of a conserved motif that directs efficient uptake of the phages. A large proportion of the peptide motifs thus obtained obeys to the proprotein convertase consensus R-X-X-R. This observation forms the basis of the proposal to use proprotein convertases as vehicles for the intracellular delivery of potential drugs and macromolecules to specific cell-types.
Table 5.
Cell TypeSequenceSequence SEQ
m ID
NO

1. PancreaticP1 RRTK 1 (3-Cells 2. P6 RKLR 2 3. P66 RRPK 3 4. I2 PTAKPTYTK 4 5. I6 IQGNGRQVGCLTNK 5 6. I10 MRGLSKRG 6 7. He atocytesH2 RQFRK 7 8. H4 RRIRG 8 9. H6 NRRRGIN 9 10. H16 KGKW 10 11. Colon WP2 RGNRGAR 11 Cancer 12. Muscles Ml RRPR 12 13. M2 GRRKG 13 14. M3 ERRK 14 15. M4 SGGRKQR 15 16. M6 RSKR 16 17. M7 RRSGR 17 18. M9 KQRR 18 19. Mll GKRAR 19 20. M13 TGKRM__T_R 20 21. _ A2 KRGR 21 Lung 22. A3 SLRRR 22 23. A8 PSLRRPR 23 24. A10 YKRGR 24 25. A16 GMGRKPR 25 26. T1 RRRVG 26 27. T2 RSFGVKKYG 27 28. T3 KSLRSFK 28 29. TS RVRR 29 30. T7 PRSRR 30 31. T8 MRRR 31 32. T10 YGGKRTLAMSK 32 33. T11 GRRSR 33 34. T13 YPLPNMK 34 Example IV: Multimeric Conjugates.
Ligand peptides obtained from phage library can be of low affinity (micromolar range).
Their binding affinity or avidity may be enhanced by multimerizing copies of the peptide molecule, e.g., by generating a peptabody. See Scott and Smith, Science 249:386-90 (1990);
Renschler et al., Proc. Natl. Acad. Sci. USA 91:3623-3627 (1994); Alexey et al., Proc. Natl.
Acad. Sci. USA 94:1663-1668 (1997). Phage experiments indicated that the phage peptide RRTK (SEQ ID NO:1, see above) was taken up specifically by (3TC-3 cells via receptor-mediated endocytosis, although the efficiency of internalization was low (which might be due to its low-affinity binding (approximately micromolar range). A series of multimers (FIGS l and 2) displaying multiple copies (four or eight) of this peptide along with a reporter tag (either FITC or biotin) was constructed.
Multimeric transport peptides of the invention can be represented by Formula I:
[P]~ [Spacer]X Core-[Linker]y-[Reporter]Z
where P is a transporter peptide, such as the consensus motifs (XmRXoRXn)i (XmRRRXn); (XmRWXRXn); and (XmRXRRXX"), as described above. For example, the peptide a peptide can include the sequence Xl-X2~3-X4, where Xl and X4 can be R or K and Xl and X4 can be any amino acid. The number of peptides in a given transporter peptide, represented by v, is an integer from two to eight or more. In any given multimeric transporter peptide, each of the peptides may be the same as or different from each other. Preferred peptides include those listed in Table 5.
A spacer may be present or absent between the peptide and core, thus x is one or zero.
Preferred spacers are flexible, amphiphilic, non-immunogenic and unsusceptible to proteases, and include polyethylene glycols (PEG) such as succinimidyl-PEG (succ-peg).
The core moieties contemplated include the polylysine (K) core described in Table 7 and include those structures having a built-in linker. The core chosen determines the number of peptides present within a given conjugate. Typical cores include: C4-KaK-K(succ-peg-S)-amide;
C-GGG -[K(C)]3-K(succ-peg-S)-amide; (NH20CHZC0)4-K2K-K(succ-peg)-amide;
(NHaOCHZCO)8-KaKaK-K(GGG)-amide. The linker and reporter groups can be present or absent, thus y and z are, independently, one or zero in Formula I.
The linker can be present or absent (i.e., y is one or zero). Linkers include those moieties which connect a core to a reporter and can be, for example, succ-PEG or NHaOCH~CO-Lys.
The reporter group can be present or absent (z is one or zero). The reporter group is any group capable of detection. Non-limiting examples include radioactive isotopes, fluorescent moieties, phosphorescent moieties, chemiluminescent moieties, and quantum dots. Other reporter groups include biotin, cysteine, histidine, haemagglutinin, myc or flag tags. A typical fluorescent reporter group used herein is FITC (fluorescein isothiocyanate).
Representative multimeric transporter peptides are shown in FIGs 1 and 2.
In a multimeric transporter peptide unit or conjugate, the multimeric transporter peptide is fused to one or more effectors. The effector can be any suitable molecule, including DNA, RNA, a protein, a peptide, or a pharmaceutically active agent, such as, for example, a toxin, an antibiotic, an antipathogenic agent, an antigen, an antibody, an antibody fragment, an immunomodulator, an enzyme, or a therapeutic agent. When more than one effector is present, the effectors can be the same or different.
Thus, the multimeric conjugates of the invention can be represented by Formula (II):
([P],; [Spacer]X Core-[Linker]y-[Reporter]~)~E
where P, Core, Spacer, Linker, Reporter, v, x, y, and z are as described above and E is an effector. In a covalently fused conjugate, the effector can be attached to the core, to the linker, or to both, as allowed by the laws of chemical bonding.
Peptide synthesis The peptide sequence RRTK (P1, SEQ ID N0:1) was obtained from the phage display experiments that showed the peptide consensus motif Xl-XZ_X3-X4, where Xl and X4 can be R or K and Xl and X4 can be any amino acid, for binding to pancreatic [3-cells.
Peptides modified as seen in Table 6 were synthesized manually on 0.5 mmol Boc-Lys(CIZ)-PAM resin by Boc chemistry. Italic type denotes linker residues and space not present in the corresponding protein sequences. Several core structures used in this experiment are shown in Table 7. Synthesis was performed on 0.5 mmol methylbenzhydrylamine resin (MBHA resin) using Boc chemistry.
All peptides were cleaved from the resin with HF (0 °C for 1 h in the presence of 5%
p-cresol), precipitated with cold ether, extracted with 50% acetonitrile, filtered, and lyophilized.
The crude peptides were purified by preparative HPLC and characterized by ESIMS.
Table 6: Synthetic peptide sequences modified from RRTK (SEQ
ID NO:1) Peptide 1 ~ BrCH2C0-RRTK-OH

Peptide 2 BrCHZCO-(peg-succ)2-RRTK-OH

Peptide 3 H S-(pog-succ)2-RRTK-OH (L-form) Peptide 4 H S (pog-suce)Z-RRTK-OH (D-form) Table 7: Core structures used Core 1 C4-KZK-K(succ-peg-S)-amide Core 2 C-GGG -[K(C)]3-K(succ-peg-S)-amide Core 3 (NH20CHZC0)4-KZK-K(succ-peg-Biotin)-amide Core 4 (NHzOCHaCO)$-K4KzK-K(GGG-Biotin)-amide Preparation of a Fluoresceira linker A fluorescein linker NH20CHaC0-Lys(FITC)-OH was prepared according to Offord's method. See Offord, et al., Methods Enzymol. 287:348-69 (1997). In brief, 2 mmol of Boc-aminooxyacetyl (Boc-AOA-OSu) and 1.2 mmol of NE-(TFA)-Lys were added into 3 ml of DMSO (dimethylsulfoxide) solution, and N-Ethyhnorpholine was added until pH
value 8-9.
After incubation at room temperature for 15 hours and then at 37 °C for 1 hour, the pH value was brought to 3.0 by careful addition of glacial acetic acid under constant agitation. The product [Boc-AOA-Lys(TFA)-OH] was isolated on a preparative-scale HPLC and then dried.
4 ml water was added and then 0.44 ml piperidine (final concentration 1 M) into the dried compound, which was incubated 4 hours at room temperature, followed by pH adjustment to 3.0 on ice. The mixture was then diluted with 10 ml of 0.1% TFA. The deprotected material (Boc-AOA-Lys-OH) was isolated by preparative HPLC and dried again. The dried material was dissolved in 300 p,l N,N-dimethylformamide (DMF), 40 mg fluorescein isothiocyanate (FITC) was added, and the mixture was adjusted to pH 8.0 with N-ethylinorpholine. After incubation for 15 hours in the dark, the product was purified by chromatography on a silica column [Kieselgel 60 (Fluka Chemie, Buchs, Switzerland), 1.5 x 20 cm] equilibrated in methanol/CH3Cl (1:1, v/v). The excess FITC eluted in the flow-through fraction, and a second yellow fraction containing the expected product was eluted with methanol/CH3-Cl (4:1, v/v). The solvent was removed by rotary evaporation, and the dried compound, Boc-AOA-Lys(FITC)-OH, was deprotected in TFA
(at a concentration not exceeding 20 mg/ml) and left for 45 min at room temperature. The majority of the TFA was then evaporated and drying was completed by lyophilization. The final product [AOA-Lys(FITC)-OH] was isolated by preparative HPLC and characterized by ESI-MS.
Alkylatioia of peptide ligahds to the core 0.9 pmol of Cys-core peptide (1.19 mg) was dissolved in 50 p,l of acetonitrile and 100 pl of water (peptide not fully soluble). A fresh solution of bromoacetyl-peptide was prepared (3.6 ~.mol in O.1M sodium phosphate buffer, pH 7.0). Then the alkylation was started by mixing two solutions at room temperature in dark. After 40 min, the product was purified on semi-preparative RP-HPLC, lyophilized, and characterized.
Periodate oxidation For periodate oxidation, 0.375 pmol of peptide (peptides 3 or 4 from Table 6, or the alkylated peptides listed in Table 8) was dissolved in 433 ~.1 acetonitrile and 1.26 ml imidazole buffer (50 mM, pH 6.95, chloride counter ion). Methionine (113 ~,1, 200 mM) was added. Then 17 p,l of Na04 (0.1 M in water) was added to start the oxidation. After 5 minutes, the reaction was quenched by adding 40 p,l of ethylene glycol (500 mM in water). The product was purified on preparative HPLC and characterized by mass spectrometry.
Table 8:
Alkylated peptides*

1 H-S-(peg-succ)2-RRTK-OH

2 H-S-(peg-succ)2-RRTK-OH (D-form) 3 [HO-KTRR-COCH2-S-C]4-K2K(succ-peg-S)-amide 4 [HO-KTRR-(succ-peg)2-COCHa-S-C]4-KZK(succ-peg-S)-amide 5 HO-KTRR-COCH2-S-C-G3-[K(C-S-CHZCO-RRTK OH)]3-K(succ-peg-S)-amide *A bold letter denotes an amino acid residue.
Oximation The aldehyde obtained by periodate oxidation was then reacted with core 3 or 4 (containing an AOA group in each core), or AOA-Lys(FITC)-OH, to form an oxime.
Oximes 1 -6, shown in FIGS l and 2, were generated in this manner. The expected and found Mass Spec data are also provided in Table 9. As shown in FIG. 1, for oximes 1 - 3, the branching Lys residues of the cores were acylated on both alpha and epsilon amino groups with the N-terminus of RRTK (SEQ ID NO: l) peptide with or without a spacer. A reporter tag, FITC, together with a spacer was introduced into the core of each construct. In FIG. 2, oximes 4 -6, the branching Lys residues of the cores were acylated on both alpha and epsilon amino groups with the N-terminus of RRTK(SEQ ID NO:l) peptide together with a spacer. A reporter tag, biotin, together with a spacer was introduced into the core of each construct.
Table 9 Oximes.
Oxime ExpectedFound Mass Mass S ec S ec Oxime 4289.04 4289.31 Oxime 4460.18 4459.48 Oxime 6708.00 6708.29 Oxime 6160.3 6160.0 Oxime 6160.3 61 S
8.6 Oxime 11643.611646.1 For oximations with AOALys(FITC)-OH, 0.129 wmol of oxidized construct was dissolved in 5 ~.1 of acetonitrile and 200 ~,l of NaOAc buffer (acetic acid 0.57 ml in 100 ml of water, sodium acetate 0.82 g dissolved in 100 ml of water, mixed part of both until pH 4.0).
5 0.516 wmol AOA-Lys(FITC)-OH (8.8 mg/ml in 100 pl acetonitrile and 100 ~.l of NaOAc buffer) was added into the mixture, followed by addition of acetic acid to a final concentration of 4%.
For oximation with core 3 or 4, 3.8 p,mol of oxidized peptide 3 or 4 was dissolved in 100 pl of acetonitrile and 200 pl of NaOAc buffer. 0.475 p,mol core 3 or 0.237 wmol core 4 was added into the solution, followed by addition of acetic acid to a final concentration of 4%. After 24 h reaction at room temperature, the product was purified on semi-preparative RP-HPLC and characterized by mass spectrometry.
Oxime 1 (FIG. 1) was constructed on a radial trilysine core (core 1) possessing a copy of RRTK (SEQ ID NO:l) peptide (peptide 1) at the end of each of 4 branches. Each peptide was N-terminally conjugated through a thioether bond arising from a specific reaction between bromoacetyl- and thiol- groups. The trilysine core also contained a succinimidyl-PEG spacer and Serine capable of undergoing oxidation to generate an aldehyde, and thus the reporter group aminooxyacetyl-Lys(FITC) was introduced into the tetramer via oxime formation.
HPLC and ESI-MS analysis revealed the desired product: Mass Spec data: Mr found 4289.31, Mr theoretical 4289.04. Oxime 1 was stable at acidic and neutral pH over 24 h.
Oximes 2 and 3 (FIG. 1) were similarly formed, however, on oxime 3 a two-unit PEG
spacer was placed between the RRTK peptide (SEQ ID NO:1) and the core, and on oxime 2 the RRTK peptide (SEQ ID NO:1) was N-terminally conjugated to core 2 instead of core 1. Mass Spec data: oxime 2: Mr found 4459.48, Mr theoretical 4460.19; oxime 3: Mr found 6708.29, Mr theoretical 6708.00.
Oxime 4 (FIG. 2) was synthesized similarly to oxime 3, but a biotin group was attached directly via amide bond on the trilysine core, instead of using FITC group as a reporter. Mass Spec data: Mr found 6160.0, Mr theoretical 6160.3. oxime 5 was similar to Oxime 4, but the RRTK (SEQ ID NO:1) residues were all D-form. Mass Spec data: Mr found 6158.6, Mr theoretical 6160.3.
Oxime 6 was synthesized as oxime 4, but it contains eight copies of RRTK(SEQ m NO:1) peptide instead of four copies. Mass Spec data: Mr found 11646.1, Mr theoretical 11643.6.
ImrnurZOCytochernistry and fluorescence studies Single phages isolated according to the enrichment scheme above were amplified. The phage peptides and one of oximes 1-6 were added and incubated for 24 hours with cells in culture medium. Medium was then washed off and cells fixed in cold methanol-acetone (l :l) for 5 minutes. Antibodies directed against the phage capsid or against biotin (peptides) were used with a fluorescein-conjugated (phage) or Texas Red labeled (peptides) secondary antibody.
Classical fluorescence microscopic studies and confocal microscopic assays were performed.
Oximes 1-3 were first tested for their entry into (3TC-3 cells. Fluorescence studies showed that oxime 3, a tetraoxime (tree design) employing four copies of RRTK
peptide with 2 units of PEG space as building blocks, clearly bound to the receptors on the surface of (3TC-3 cells and was able to enter into the cells. The superiority of oxime 3 may be due to the flexibility of the PEG spacer, which increases binding to the receptors on the cells.
Oximes 4-6, which are similar to oxime 3, were then synthesized and assayed for binding avidity and entry efficiency. The number of copies of the RRTK(SEQ ID NO:l) peptide was increased up to eight in oxime 6, and, because D-peptides had been demonstrated to possess an increased stability and also lower immunogenicity (See Ivanenkov et al., Biochem. Biophys.
Acta 1448:450 (1999)), the peptide was changed into its D- enantiomeric form in oxime 5:
Microscopic examination showed that oxime 6, with eight copies of peptide (octaoxime), was able to efficiently enter cells. Texas Red labeled secondary antibody to the Biotin demonstrated that the octaoxime localized either inside cells as "vacuolar" structures, or at the cell membrane.
Therefore, multimers where the peptide copy number is increased up to eight, exhibit enhanced avidity of the binding peptides and transport efficiency.

Claims (46)

1. A multimeric transporter peptide according to Formula I, [P]v-[Spacer]x-Core-[Linker]y-[Reporter]z (I) wherein P is a transporter peptide, comprising at least one amino acid sequence selected from the group consisting of:

a) (X m RX o RX n);
b) (X m RRRX n);
c) (X m RRXRX n); and d) (X m RXRRX n), wherein X is a non-basic amino acid;
m is an integer from zero to fourteen;
n is an integer, independent of m, between zero and fourteen;
o is an integer, independent of m and n, between zero and five;
v is an integer from two to eight;
x, y and z, are, independently, zero or one; and wherein each P can be the same or different, and said transporter peptide is capable of translocating across a biological membrane.
2. The transporter peptide of claim 1, wherein at least one P has the amino acid sequence R-X-X-R.
3. The transporter peptide of claim 1, wherein each P is selected from the group consisting of SEQ ID NOs: 1-34.
4. The transporter peptide of claim 1, wherein v is selected from the group consisting of two, four, six, and eight.
5. The transporter peptide of claim 4, wherein v is four.
6. The transporter peptide of claim 4, wherein v is eight.
7. The transporter peptide of claim 1, wherein the Spacer is succinimidyl-polyethylene glycol.
8. The transporter peptide of claim 1, wherein the Core is selected from the group consisting of C4-K2K-K(succ-peg-S)-amide; C-GGG -[K(C)]3-K(succ-peg-S)-amide;
(NH2OCH2CO)4-K2K-K(succ-peg)-amide; and (NH2OCH2CO)8-K4K2K-K(GGG)-amide.
9. A transporter unit comprising the transporter peptide of claim 1 conjugated to an effector.
10. A transporter unit of Formula II:

([P]v-[Spacer]x-Core-[Linker]y-[Reporter]z).cndot.E (I) wherein: E is an effector;
P is a transporter peptide, comprising at least one amino acid sequence selected from the group consisting of:
e) (X m RX o RX n);
f) (X m RRRX n);
g) (X m RRXRX n); and h) (X m RXRRX n), wherein X is a non-basic amino acid;
m is an integer from zero to fourteen;
n is an integer, independent of m, between zero and fourteen;
o is an integer, independent of m and n, between zero and five;
v is an integer from two to eight;
x, y and z, are, independently, zero or one; and wherein each P can be the same or different, and wherein said transporter peptide is capable of translocating across a biological membrane.
11. The transporter unit of claim 10, wherein at least one P has the amino acid sequence R-X-X-R.
12. The transporter unit of claim 10, wherein each P is selected from the group consisting of SEQ ID NOs: 1-34.
13. The transporter unit of claim 10, wherein v is selected from the group consisting of two, four, six, and eight.
14. The transporter unit of claim 13, wherein v is four.
15. The transporter unit of claim 13, wherein v is eight.
16. The transporter unit of claim 10, wherein the core is selected from the group consisting of C4-K2K-K(succ-peg-S)-amide; C-GGG -[K(C)]3-K(succ-peg-S)-amide; (NH2OCH2CO)4-K(succ-peg)-anode; and (NH2OCH2CO)8-K4K2K-K(GGG)-amide.
17. The transporter unit of claim 10, wherein the effector is covalently fused to the linker.
18. The transporter unit of claim 10, wherein the effector is selected from the group consisting of a nucleic acid; a peptide; and a pharmaceutically active agent.
19. The transporter unit of claim 18, wherein said effector is a nucleic acid.
20. The transporter unit of claim 19 wherein said nucleic acid is DNA.
21. The transporter unit of claim 19, wherein said nucleic acid is RNA.
22. The transporter unit of claim 18, wherein said effector is a peptide.
23. The transporter unit of claim 18, wherein said effector is a pharmaceutically active agent.
24. The transporter unit of claim 23, wherein said pharmaceutically active agent is selected from the group consisting of a toxin, an antibiotic, an antipathogenic agent, an antigen, an antibody fragment, an immunomodulator, an enzyme, and a therapeutic agent.
25. The transporter unit of claim 10 wherein each peptide is less than 50 amino acids long.
26. The transporter unit of claim 10 wherein each peptide is less than 25 amino acids long.
27. The transporter unit of claim 10 wherein each peptide is less than 15 amino acids long.
28. The transporter unit of claim 10 wherein translocation occurs within a tissue selected from the group consisting of pancreatic B-cells; hepatocytes; colon cells;
muscle cells; and lung cells.
29. A method of translocating the transporter unit of claim 12 across the membrane of pancreatic .beta.-cells, wherein at least one transporter peptide is selected from the group consisting of SEQ ID NOS: 1-6.
30. The method of claim 29, wherein at least one transporter peptide is SEQ ID
NO: 1.
31. A method of translocating the transporter unit of claim 12 across the membrane of hepatocytes, wherein at least one peptide is selected from the group consisting of SEQ ID NOS:
7-10.
32. A method of translocating the transporter unit of claim 12 across the membrane of colon cells, wherein at least one peptide is SEQ ID NO: 11.
33. A method of translocating the transporter unit of claim 12 across the membrane of muscle cells, wherein at least one peptide is selected from the group consisting of SEQ ID NOS: 12-20.
34. A method of translocating the transporter unit of claim 12 across the membrane of lung cells, wherein at least one peptide is selected from the group consisting of SEQ ID NOS: 21-34.
35. A pharmaceutical composition comprising a therapeutically or prophylactically effective amount of the transporter unit according to claim 10, and a pharmaceutically acceptable carrier.
36. A method of producing a translocatable conjugate between the transporter peptide of claim 1 and an effector, said method comprising conjugating said effector to said transporter peptide to form a transporter peptide-effector conjugate.
37. A method of translocating an effector into the cytoplasm and nucleus of a eukaryotic cell, said method comprising:
conjugating said effector to the transporter peptide of claim 1 to form a transporter peptide-effector conjugate; and introducing said transporter peptide-effector conjugate to the cell.
38. The method of claim 37, wherein the introducing step is achieved by incubating a cell culture in the presence of said transporter peptide-effector conjugate or by injecting said transporter peptide-effector conjugate into the cell.
39. The method of claim 37, wherein the eukaryotic cell is a human cell.
40. The method of claim 37, wherein the eukaryotic cell is a .beta.-cell.
41. A method of increasing the intracellular concentration of an effector within a eukaryotic cell, said method comprising;
conjugating said effector to the transporter peptide of claim 1 to form a transporter peptide-effector conjugate;
incubating said cell in the presence of said transporter peptide-effector conjugate, under conditions promoting active metabolism of said eukaryotic cell.
42. The method of claim 41, wherein the eukaryotic cell is a human cell.
43. A kit comprising in one or more containers, a therapeutically or prophylactically effective amount of the pharmaceutical composition of claim 35.
44. A method of treating or preventing a disease, said method comprising administering to a subject in which such treatment or prevention is desired the pharmaceutical composition of claim 35, in an amount sufficient to treat or prevent said disease in said subject.
45. The method of claim 44, wherein said disease is selected from a group consisting of diabetes, colon cancer, respiratory ailments, neurodegenerative disorders, cardiplegia, and viral infections.
46. A multimeric transporter peptide, wherein each transporter peptide amino acid sequence is selected from the group consisting of SEQ ID NOS: 1-34.
CA002488716A 2002-06-07 2003-06-06 Intracellular delivery of biological effectors Abandoned CA2488716A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US10/165,015 US7033597B2 (en) 2000-10-13 2002-06-07 Intracellular delivery of biological effectors
US10/165,015(CIP) 2002-06-07
PCT/IB2003/003097 WO2003103718A2 (en) 2002-06-07 2003-06-06 Intracellular delivery of biological effectors

Publications (1)

Publication Number Publication Date
CA2488716A1 true CA2488716A1 (en) 2003-12-18

Family

ID=29732051

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002488716A Abandoned CA2488716A1 (en) 2002-06-07 2003-06-06 Intracellular delivery of biological effectors

Country Status (6)

Country Link
US (2) US7033597B2 (en)
EP (1) EP1511763A2 (en)
JP (2) JP4514607B2 (en)
AU (1) AU2003274822B2 (en)
CA (1) CA2488716A1 (en)
WO (1) WO2003103718A2 (en)

Families Citing this family (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8183339B1 (en) 1999-10-12 2012-05-22 Xigen S.A. Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US20040082509A1 (en) 1999-10-12 2004-04-29 Christophe Bonny Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US20040248158A1 (en) * 2003-01-28 2004-12-09 Loughran Thomas P Differentially expressed genes in large granular lymphocyte leukemia
CA2516685A1 (en) 2003-02-20 2004-09-02 University Of South Florida Peptidomimetic inhibitors of stat3 activity and their medical uses
WO2004094463A2 (en) * 2003-04-18 2004-11-04 University Of Florida Research Foundation, Inc. Peptide inhibitors of autophosphorylation protein kinases
EP1664070B1 (en) * 2003-08-13 2008-08-13 University Of South Florida Methods for inhibiting tumor cell proliferation involving platinum complexes
US8227434B1 (en) 2003-11-04 2012-07-24 H. Lee Moffitt Cancer Center & Research Institute, Inc. Materials and methods for treating oncological disorders
US20050288365A1 (en) * 2004-01-06 2005-12-29 Heidi Kay Platinum complexes and methods for inhibiting tumor cell proliferation
US7625885B2 (en) * 2004-02-17 2009-12-01 University Of South Florida Cytotoxin compound and method of isolation
US8669376B2 (en) * 2004-02-17 2014-03-11 University Of South Florida Cytotoxin compounds and methods of isolation
WO2007031098A1 (en) 2005-09-12 2007-03-22 Xigen S.A. Cell-permeable peptide inhibitors of the jnk signal transduction pathway
US8080517B2 (en) 2005-09-12 2011-12-20 Xigen Sa Cell-permeable peptide inhibitors of the JNK signal transduction pathway
WO2007047623A2 (en) * 2005-10-14 2007-04-26 University Of South Florida Methods of inhibiting cell growth and methods of enhancing radiation responses
DE102006040647A1 (en) * 2006-08-30 2008-03-13 Robert Bosch Gmbh Hand tool
WO2008079945A2 (en) * 2006-12-20 2008-07-03 University Of South Florida Rock inhibitors and uses thereof
ES2668537T3 (en) * 2007-07-13 2018-05-18 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Antimicrobial peptides derived from viruses
WO2009143864A1 (en) 2008-05-30 2009-12-03 Xigen S.A. Use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of chronic or non-chronic inflammatory digestive diseases
WO2009143865A1 (en) 2008-05-30 2009-12-03 Xigen S.A. Use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of various diseases
US8673910B2 (en) * 2008-06-30 2014-03-18 H. Lee Moffitt Cancer Center And Research Institute Proteasome inhibitors for selectively inducing apoptosis in cancer cells
US9174969B2 (en) * 2008-07-21 2015-11-03 University Of South Florida Indoline scaffold SHP-2 inhibitors and cancer treatment method
US9132168B2 (en) 2008-08-05 2015-09-15 University Of South Florida Methods of treating cognitive impairment
JP2011530520A (en) * 2008-08-05 2011-12-22 ユニバーシティー オブ サウス フロリダ How to treat cognitive impairment
WO2010072228A1 (en) * 2008-12-22 2010-07-01 Xigen S.A. Novel transporter constructs and transporter cargo conjugate molecules
WO2011008863A2 (en) * 2009-07-14 2011-01-20 Lucia Irene Gonzalez Stereoisomer peptides and their polymer conjugates for hiv disease
US9221808B2 (en) 2010-04-16 2015-12-29 H. Lee Moffitt Cancer Center And Research Institute, Inc. Pyridylthiazole-based ureas as inhibitors of Rho associated protein kinase (ROCK) and methods of use
WO2011160653A1 (en) 2010-06-21 2011-12-29 Xigen S.A. Novel jnk inhibitor molecules
US9150618B2 (en) 2010-10-14 2015-10-06 Xigen Inflammation Ltd. Use of cell-permeable peptide inhibitors of the JNK signal transduction pathway for the treatment of chronic or non-chronic inflammatory eye diseases
WO2012135697A2 (en) 2011-03-30 2012-10-04 H. Lee Moffitt Cancer Center & Research Institute Inc. Novel rho kinase inhibitors and methods of use
WO2013091670A1 (en) 2011-12-21 2013-06-27 Xigen S.A. Novel jnk inhibitor molecules for treatment of various diseases
WO2013112722A1 (en) 2012-01-24 2013-08-01 H. Lee Moffitt Cancer Center And Research Institute, Inc. Inhibitors of rho associated protein kinases (rock) and methods of use
CA2866707A1 (en) 2012-03-07 2013-09-12 H. Lee Moffitt Cancer Center And Research Institute, Inc. Selective histone deactylase 6 inhibitors
US10098967B2 (en) 2012-12-03 2018-10-16 Ohio State Innovation Foundation Self-assembly of therapeutic agent-peptide nanostructures
TR201905612T4 (en) 2012-12-05 2019-05-21 Univ Heidelberg Ruprecht Karls Conjugates of proteins and polyvalent cell-penetrating peptides and their uses.
WO2014140113A1 (en) * 2013-03-13 2014-09-18 Biocrine Ab Methods for treating and/or limiting development of diabetes
CA2903275A1 (en) 2013-06-26 2014-12-31 Xigen Inflammation Ltd. New use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of various diseases
WO2014206427A1 (en) 2013-06-26 2014-12-31 Xigen Inflammation Ltd. New use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of various diseases
WO2015197097A1 (en) 2014-06-26 2015-12-30 Xigen Inflammation Ltd. New use for jnk inhibitor molecules for treatment of various diseases
WO2015021149A1 (en) 2013-08-06 2015-02-12 H. Lee Moffitt Cancer Center And Research Institute, Inc. Inhibitors of ack1/tnk2 tyrosine kinase
US10626178B2 (en) 2013-08-21 2020-04-21 Raz Yirmiya Treatment of mood and stress related disorders
US9950036B2 (en) 2013-08-21 2018-04-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Treatment of mood and stress related disorders
EP3097088A4 (en) 2014-01-23 2017-10-04 H. Lee Moffitt Cancer Center And Research Institute, Inc. Icariin derivatives
WO2015112904A1 (en) 2014-01-24 2015-07-30 University Of Florida Research Foundation, Inc. Socs mimetics for the treatment of diseases
WO2015116782A1 (en) 2014-01-29 2015-08-06 Board Of Regents, The University Of Texas System Nucleobase analogue derivatives and their applications
US10197575B2 (en) 2014-03-31 2019-02-05 H. Lee Moffitt Cancer Center And Research Institute, Inc. Stabilized peptoid-peptide hybrids and uses thereof
US9987258B2 (en) 2014-04-06 2018-06-05 H. Lee Moffitt Cancer Center And Research Institute, Inc. Histone deacetylase as a modulator of PDL1 expression and activity
WO2015157636A1 (en) 2014-04-10 2015-10-15 H. Lee Moffitt Cancer Center And Research Institute, Inc. Enhanced expansion of tumor-infiltrating lymphocytes for adoptive cell therapy
CN106852146B (en) 2014-05-21 2021-08-13 恩特拉达治疗学股份有限公司 Cell penetrating peptides and methods of making and using the same
US10815276B2 (en) 2014-05-21 2020-10-27 Entrada Therapeutics, Inc. Cell penetrating peptides and methods of making and using thereof
WO2016033368A1 (en) 2014-08-27 2016-03-03 Ohio State Innovation Foundation Improved peptidyl calcineurin inhibitors
US10118942B2 (en) 2015-03-26 2018-11-06 H. Lee Moffitt Cancer Center And Research Institute, Inc. Toll-like receptor 2 ligands and methods of making and using thereof
JP6959142B2 (en) 2015-05-06 2021-11-02 エイチ リー モフィット キャンサー センター アンド リサーチ インスティテュート インコーポレイテッド Radiation therapy agents and companion imaging agents targeting MC1R
CN106714836A (en) 2015-06-05 2017-05-24 H·李·莫菲特癌症中心研究有限公司 Gm-csf/cd40l vaccine and checkpoint inhibitor combination therapy
CN108348779A (en) 2015-08-02 2018-07-31 H·李·莫菲特癌症中心研究有限公司 The inhibitor of ACK1/TNK2 tyrosine kinase
CN108884124A (en) 2015-09-17 2018-11-23 俄亥俄州创新基金会 Caborane compounds and its application method
WO2017066428A1 (en) 2015-10-13 2017-04-20 H. Lee Moffitt Cancer Center & Research Institute, Inc. Brd4-kinase inhibitors as cancer therapeutics
AU2016366529A1 (en) 2015-12-09 2018-06-07 Vanderbilt University Biosynthesis of everninomicin analogs in Micromonospora carbonacea var aurantiaca
US11203576B2 (en) 2016-03-11 2021-12-21 H. Lee Moffitt Cancer Center And Research Institute, Inc. Aurora kinase and Janus kinase inhibitors for prevention of graft versus host disease
MX2018011216A (en) 2016-03-16 2019-08-29 H Lee Moffitt Cancer Ct & Res Small molecules against cereblon to enhance effector t cell function.
US10426843B2 (en) 2016-05-04 2019-10-01 H. Lee Moffitt Cancer Center And Research Institute, Inc. Delta-opioid receptor targeted agent for molecular imaging and immunotherapy of cancer
JP7175887B2 (en) 2016-11-09 2022-11-21 オハイオ・ステイト・イノベーション・ファウンデーション Disulfide-containing cell membrane penetrating peptide and method for producing and using same
WO2018098282A2 (en) 2016-11-22 2018-05-31 Ohio State Innovation Foundation Cyclic cell penetrating peptides comprising beta-hairpin motifs and methods of making and using thereof
WO2018098226A1 (en) 2016-11-22 2018-05-31 Ohio State Innovation Foundation Bicyclic peptidyl inhibitor of tumor necrosis factor-alpha
US11583525B2 (en) 2017-06-06 2023-02-21 H. Lee Moffitt Cancer Center And Research Institute, Inc. Geranylgeranyltransferase I inhibitor for treatment of a PTEN defective cancer
CA3078504A1 (en) 2017-10-04 2019-04-11 Ohio State Innovation Foundation Bicyclic peptidyl inhibitors
WO2019084528A1 (en) 2017-10-27 2019-05-02 Ohio State Innovation Foundation Polypeptide conjugates for intracellular delivery of stapled peptides
EP3749365A4 (en) 2018-01-29 2022-01-12 Ohio State Innovation Foundation Peptidyl inhibitors of calcineurin-nfat interaction
EP3755351A4 (en) 2018-02-22 2021-11-24 Entrada Therapeutics, Inc. Compositions and methods for treating mitochondrial neurogastrointestinal encephalopathy
EP3787671A4 (en) 2018-05-04 2022-06-15 Ohio State Innovation Foundation Non-peptidic cell-penetrating motifs
US11730726B2 (en) 2018-07-11 2023-08-22 H. Lee Moffitt Cancer Center And Research Institute, Inc. Dimeric immuno-modulatory compounds against cereblon-based mechanisms
US10898460B1 (en) 2018-07-20 2021-01-26 University Of South Florida Leishmania inhibitors
JP2023501385A (en) 2019-11-08 2023-01-18 ユニバーシティ オブ ピッツバーグ - オブ ザ コモンウェルス システム オブ ハイヤー エデュケイション Compositions containing antimicrobial peptides
US20230190871A1 (en) 2020-05-20 2023-06-22 Sana Biotechnology, Inc. Methods and compositions for treatment of viral infections
IL307298A (en) 2021-03-31 2023-11-01 Entrada Therapeutics Inc Cyclic cell penetrating peptides
WO2022240721A1 (en) 2021-05-10 2022-11-17 Entrada Therapeutics, Inc. Compositions and methods for modulating interferon regulatory factor-5 (irf-5) activity
EP4337261A2 (en) 2021-05-10 2024-03-20 Entrada Therapeutics, Inc. Compositions and methods for modulating mrna splicing
JP2024518068A (en) 2021-05-10 2024-04-24 エントラーダ セラピューティクス,インコーポレイティド Compositions and methods for intracellular therapy
KR20240038967A (en) 2021-06-23 2024-03-26 엔트라다 테라퓨틱스, 인크. Antisense compounds and methods for targeting CUG repeats
AU2022339769A1 (en) 2021-09-01 2024-03-28 Entrada Therapeutics, Inc. Compounds and methods for skipping exon 44 in duchenne muscular dystrophy
WO2023219933A1 (en) 2022-05-09 2023-11-16 Entrada Therapeutics, Inc. Compositions and methods for delivery of nucleic acid therapeutics
WO2024015924A2 (en) 2022-07-15 2024-01-18 Entrada Therapeutics, Inc. Hybrid oligonucleotides
WO2024073042A1 (en) 2022-09-30 2024-04-04 Entrada Therapeutics, Inc. Ocular delivery of therapeutic agents

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0632722A4 (en) * 1992-03-20 1997-07-30 Baylor College Medicine A dna transporter system and method of use.
WO1993019768A1 (en) * 1992-04-03 1993-10-14 The Regents Of The University Of California Self-assembling polynucleotide delivery system
CA2094658A1 (en) 1992-04-23 1993-10-24 Martin Sumner-Smith Intracellular delivery of biochemical agents
EP0693939A1 (en) 1993-04-14 1996-01-31 Roche Diagnostics GmbH Nucleic acid tranfer peptides and their use for injecting nucleic acids into eucaryotic cells
US6361938B1 (en) 1996-11-08 2002-03-26 Elan Corporation, Plc Peptides which enhance transport across tissues and methods of identifying and using the same
EP1019071A4 (en) 1997-05-15 2003-07-30 Cytogen Corp Random peptides that bind to gastro-intestinal tract (git) transport receptors and related methods
EP0975370B9 (en) * 1997-05-21 2004-11-03 The Board Of Trustees Of The Leland Stanford Junior University Composition and method for enhancing transport across biological membranes
FR2766826B1 (en) 1997-08-04 2001-05-18 Pasteur Institut VECTORS DERIVED FROM ANTIBODIES FOR TRANSFERRING SUBSTANCES IN CELLS
FR2767323B1 (en) 1997-08-12 2001-01-05 Synt Em LINEAR PEPTIDES DERIVED FROM ANTIBIOTIC PEPTIDES, THEIR PREPARATION AND THEIR USE FOR VECTORIZING ACTIVE SUBSTANCES
CN1330675A (en) 1998-08-28 2002-01-09 格莱风科学公司 Polyamide chains of precise length, methods to manufacture them and their conjugates with protein
DK1102785T3 (en) * 1999-06-07 2013-05-13 Arrowhead Res Corp Compositions for drug delivery using pH-sensitive molecules
US20030104622A1 (en) * 1999-09-01 2003-06-05 Robbins Paul D. Identification of peptides that facilitate uptake and cytoplasmic and/or nuclear transport of proteins, DNA and viruses
FR2805821B1 (en) 2000-03-01 2004-01-16 Diatos AMINO ACID SEQUENCES TO FACILITATE THE PENETRATION OF A SUBSTANCE OF INTEREST WITHIN CELLS AND / OR CELL CORES
JP4387669B2 (en) 2000-10-13 2009-12-16 ザイジェン エス.アー. Intracellular delivery of biological effectors with novel transporter peptide sequences

Also Published As

Publication number Publication date
AU2003274822A1 (en) 2003-12-22
WO2003103718A3 (en) 2004-11-25
JP4514607B2 (en) 2010-07-28
US20060178310A1 (en) 2006-08-10
EP1511763A2 (en) 2005-03-09
AU2003274822B2 (en) 2008-01-17
JP2010024250A (en) 2010-02-04
US20030032594A1 (en) 2003-02-13
US7033597B2 (en) 2006-04-25
JP2006500917A (en) 2006-01-12
WO2003103718A2 (en) 2003-12-18

Similar Documents

Publication Publication Date Title
AU2003274822B2 (en) Intracellular delivery of biological effectors
US6960648B2 (en) Intracellular delivery of biological effectors
Bourgault et al. Novel stable PACAP analogs with potent activity towards the PAC1 receptor
Jiang et al. GnRH antagonists: a new generation of long acting analogues incorporating p-ureido-phenylalanines at positions 5 and 6
Freeman et al. Microwave-assisted solid-phase aza-peptide synthesis: aza scan of a PKB/Akt inhibitor using aza-arginine and aza-proline precursors
AU723694B2 (en) Exendin analogues, processes for their preparation and medicaments containing them
Spiegel et al. Cyclic aza-peptide integrin ligand synthesis and biological activity
Corbi-Verge et al. Strategies to develop inhibitors of motif-mediated protein-protein interactions as drug leads
JPH06505486A (en) Conformationally restricted mimetics of β-turns and β-bulges and peptides containing them
Parthasarathy et al. The medicinal chemistry of therapeutic peptides: Recent developments in synthesis and design optimizations
Jinsmaa et al. Oral bioavailability of a new class of μ-opioid receptor agonists containing 3, 6-bis [Dmt-NH (CH2) n]-2 (1 H)-pyrazinone with central-mediated analgesia
CN112041330A (en) Compstatin analogs and medical uses thereof
Mezö et al. Synthesis of Gonadotropin-Releasing Hormone III Analogs. Structure− Antitumor Activity Relationships
Ciulla et al. Nature-inspired and medicinally relevant short peptides
AU2008201592B2 (en) Intracellular Delivery of Biological Effectors
JP2018504381A (en) Cell penetrating peptide
US20230287051A1 (en) Inhibitors of complement factor c3 and their medical uses
Kubi et al. Cell-penetrating and mitochondrion-targeting molecules
Einsiedel et al. Development of disulfide-functionalized peptides covalently binding G protein-coupled receptors
AU2007216926A1 (en) Intracellular Delivery of Biological Effectors by Novel Transporter Peptide Sequences
Zamudio Vázquez Synthesis, Biological Evaluation and Insights into the Mode of Action of Quinoxaline Containing Peptides
Jessop Molecular transporters for drug delivery: Guanidinium-based and nanotube transporters
Bowerman et al. Aromatic Versus Hydrophobic Contributions to Amyloid Peptide Self-Assembly

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued